Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mutat ; 42(12): 1624-1636, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34510628

RESUMEN

N-acetylglutamate synthase deficiency is an autosomal recessive urea cycle disorder caused either by decreased expression of the NAGS gene or defective NAGS enzyme resulting in decreased production of N-acetylglutamate (NAG), an allosteric activator of carbamylphosphate synthetase 1 (CPS1). NAGSD is the only urea cycle disorder that can be effectively treated with a single drug, N-carbamylglutamate (NCG), a stable NAG analog, which activates CPS1 to restore ureagenesis. We describe three patients with NAGSD due to four novel noncoding sequence variants in the NAGS regulatory regions. All three patients had hyperammonemia that resolved upon treatment with NCG. Sequence variants NM_153006.2:c.427-222G>A and NM_153006.2:c.427-218A>C reside in the 547 bp-long first intron of NAGS and define a novel NAGS regulatory element that binds retinoic X receptor α. Sequence variants NC_000017.10:g.42078967A>T (NM_153006.2:c.-3065A>T) and NC_000017.10:g.42078934C>T (NM_153006.2:c.-3098C>T) reside in the NAGS enhancer, within known HNF1 and predicted glucocorticoid receptor binding sites, respectively. Reporter gene assays in HepG2 and HuH-7 cells demonstrated that all four substitutions could result in reduced expression of NAGS. These findings show that analyzing noncoding regions of NAGS and other urea cycle genes can reveal molecular causes of disease and identify novel regulators of ureagenesis.


Asunto(s)
N-Acetiltransferasa de Aminoácidos , Hiperamonemia , Trastornos Innatos del Ciclo de la Urea , N-Acetiltransferasa de Aminoácidos/química , N-Acetiltransferasa de Aminoácidos/genética , Humanos , Hiperamonemia/genética , Intrones , Secuencias Reguladoras de Ácidos Nucleicos , Trastornos Innatos del Ciclo de la Urea/genética
2.
J Inherit Metab Dis ; 42(6): 1088-1096, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31177541

RESUMEN

Abundance of urea cycle enzymes in the liver is regulated by dietary protein intake. Although urea cycle enzyme levels rise in response to a high-protein (HP) diet, signaling networks that sense dietary protein intake and trigger changes in expression of urea cycle genes have not been identified. The aim of this study was to identify signaling pathway(s) that respond to changes in protein intake and regulate expression of urea cycle genes in mice and human hepatocytes. Mice were adapted to either HP or low-protein diets followed by isolation of liver protein and mRNA and integrated analysis of the proteomic and transcriptomic data. HP diet led to increased expression of mRNA and enzymes in amino acid degradation pathways and decreased expression of mRNA and enzymes in carbohydrate and fat metabolism, which implicated adenosine monophosphate-activated protein kinase (AMPK) as a possible regulator. Primary human hepatocytes, treated with 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) an activator of AMPK, were used to test whether AMPK regulates expression of urea cycle genes. The abundance of carbamoylphosphate synthetase 1 and ornithine transcarbamylase mRNA increased in hepatocytes treated with AICAR, which supports a role for AMPK signaling in regulation of the urea cycle. Because AMPK is either a target of drugs used to treat type-2 diabetes, these drugs might increase the expression of urea cycle enzymes in patients with partial urea cycle disorders, which could be the basis of a new therapeutic approach.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas en la Dieta/farmacología , Enzimas/genética , Urea/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Células Cultivadas , Proteínas en la Dieta/administración & dosificación , Enzimas/efectos de los fármacos , Enzimas/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
3.
Hum Mutat ; 39(4): 527-536, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29282796

RESUMEN

The ornithine transcarbamylase (OTC) gene is on the X chromosome and its product catalyzes the formation of citrulline from ornithine and carbamylphosphate in the urea cycle. About 10%-15% of patients, clinically diagnosed with OTC deficiency (OTCD), lack identifiable mutations in the coding region or splice junctions of the OTC gene on routine molecular testing. We collected DNA from such patients via retrospective review and by prospective enrollment. In nine of 38 subjects (24%), we identified a sequence variant in the OTC regulatory regions. Eight subjects had unique sequence variants in the OTC promoter and one subject had a novel sequence variant in the OTC enhancer. All sequence variants affect positions that are highly conserved in mammalian OTC genes. Functional studies revealed reduced reporter gene expression with all sequence variants. Two sequence variants caused decreased binding of the HNF4 transcription factor to its mutated binding site. Bioinformatic analyses combined with functional assays can be used to identify and authenticate pathogenic sequence variants in regulatory regions of the OTC gene, in other urea cycle disorders or other inborn errors of metabolism.


Asunto(s)
Elementos de Facilitación Genéticos , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética , Regiones Promotoras Genéticas , Sitios de Unión/genética , Regulación de la Expresión Génica , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Masculino , Mutación , Ornitina/metabolismo , Estudios Prospectivos , Estudios Retrospectivos
4.
Mol Genet Metab ; 120(3): 198-206, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28007335

RESUMEN

This study documents the disparate therapeutic effect of N-carbamyl-l-glutamate (NCG) in the activation of two different disease-causing mutants of carbamyl phosphate synthetase 1 (CPS1). We investigated the effects of NCG on purified recombinant wild-type (WT) mouse CPS1 and its human corresponding E1034G (increased ureagenesis on NCG) and M792I (decreased ureagenesis on NCG) mutants. NCG activates WT CPS1 sub-optimally compared to NAG. Similar to NAG, NCG, in combination with MgATP, stabilizes the enzyme, but competes with NAG binding to the enzyme. NCG supplementation activates available E1034G mutant CPS1 molecules not bound to NAG enhancing ureagenesis. Conversely, NCG competes with NAG binding to the scarce M792I mutant enzyme further decreasing residual ureagenesis. These results correlate with the respective patient's response to NCG. Particular caution should be taken in the administration of NCG to patients with hyperammonemia before their molecular bases of their urea cycle disorders is known.


Asunto(s)
Adenosina Trifosfato/administración & dosificación , Carbamoil-Fosfato Sintasa (Amoniaco)/química , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/tratamiento farmacológico , Glutamatos/administración & dosificación , Adenosina Trifosfato/farmacología , Animales , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/enzimología , Quimioterapia Combinada , Femenino , Glutamatos/farmacología , Humanos , Masculino , Ratones , Mutación , Medicina de Precisión , Estabilidad Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Enfermedades Raras/tratamiento farmacológico , Enfermedades Raras/enzimología
5.
Mol Genet Metab ; 119(4): 307-310, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27771289

RESUMEN

BACKGROUND: N-acetylglutamate synthase (NAGS) plays a key role in the removal of ammonia via the urea cycle by catalyzing the synthesis of N-acetylglutamate (NAG), the obligatory cofactor in the carbamyl phosphate synthetase 1 reaction. Enzymatic analysis of NAGS in liver homogenates has remained insensitive and inaccurate, which prompted the development of a novel method. METHODS: UPLC-MS/MS was used in conjunction with stable isotope (N-acetylglutamic-2,3,3,4,4-d5 acid) dilution for the quantitative detection of NAG produced by the NAGS enzyme. The assay conditions were optimized using purified human NAGS and the optimized enzyme conditions were used to measure the activity in mouse liver homogenates. RESULTS: A low signal-to-noise ratio in liver tissue samples was observed due to non-enzymatic formation of N-acetylglutamate and low specific activity, which interfered with quantitative analysis. Quenching of acetyl-CoA immediately after the incubation circumvented this analytical difficulty and allowed accurate and sensitive determination of mammalian NAGS activity. The specificity of the assay was validated by demonstrating a complete deficiency of NAGS in liver homogenates from Nags -/- mice. CONCLUSION: The novel NAGS enzyme assay reported herein can be used for the diagnosis of inherited NAGS deficiency and may also be of value in the study of secondary hyperammonemia present in various inborn errors of metabolism as well as drug treatment.


Asunto(s)
N-Acetiltransferasa de Aminoácidos/genética , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Hiperamonemia/diagnóstico , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Acetilcoenzima A/metabolismo , N-Acetiltransferasa de Aminoácidos/metabolismo , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/deficiencia , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatología , Hígado/enzimología , Ratones , Ratones Noqueados , Espectrometría de Masas en Tándem , Trastornos Innatos del Ciclo de la Urea/genética , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/fisiopatología
6.
Int J Mol Sci ; 16(8): 18836-64, 2015 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-26274952

RESUMEN

Enzymes in the transcarbamylase family catalyze the transfer of a carbamyl group from carbamyl phosphate (CP) to an amino group of a second substrate. The two best-characterized members, aspartate transcarbamylase (ATCase) and ornithine transcarbamylase (OTCase), are present in most organisms from bacteria to humans. Recently, structures of four new transcarbamylase members, N-acetyl-L-ornithine transcarbamylase (AOTCase), N-succinyl-L-ornithine transcarbamylase (SOTCase), ygeW encoded transcarbamylase (YTCase) and putrescine transcarbamylase (PTCase) have also been determined. Crystal structures of these enzymes have shown that they have a common overall fold with a trimer as their basic biological unit. The monomer structures share a common CP binding site in their N-terminal domain, but have different second substrate binding sites in their C-terminal domain. The discovery of three new transcarbamylases, l-2,3-diaminopropionate transcarbamylase (DPTCase), l-2,4-diaminobutyrate transcarbamylase (DBTCase) and ureidoglycine transcarbamylase (UGTCase), demonstrates that our knowledge and understanding of the spectrum of the transcarbamylase family is still incomplete. In this review, we summarize studies on the structures and function of transcarbamylases demonstrating how structural information helps to define biological function and how small structural differences govern enzyme specificity. Such information is important for correctly annotating transcarbamylase sequences in the genome databases and for identifying new members of the transcarbamylase family.


Asunto(s)
Transferasas de Carboxilo y Carbamoilo/química , Transferasas de Carboxilo y Carbamoilo/genética , Secuencia de Aminoácidos , Transferasas de Carboxilo y Carbamoilo/metabolismo , Catálisis , Dominio Catalítico , Bases de Datos Genéticas , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Alineación de Secuencia , Especificidad por Sustrato
7.
Int J Mol Sci ; 16(6): 13004-22, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26068232

RESUMEN

N-acetylglutamate synthase (NAGS) catalyzes the production of N-acetylglutamate (NAG) from acetyl-CoA and L-glutamate. In microorganisms and plants, the enzyme functions in the arginine biosynthetic pathway, while in mammals, its major role is to produce the essential co-factor of carbamoyl phosphate synthetase 1 (CPS1) in the urea cycle. Recent work has shown that several different genes encode enzymes that can catalyze NAG formation. A bifunctional enzyme was identified in certain bacteria, which catalyzes both NAGS and N-acetylglutamate kinase (NAGK) activities, the first two steps of the arginine biosynthetic pathway. Interestingly, these bifunctional enzymes have higher sequence similarity to vertebrate NAGS than those of the classical (mono-functional) bacterial NAGS. Solving the structures for both classical bacterial NAGS and bifunctional vertebrate-like NAGS/K has advanced our insight into the regulation and catalytic mechanisms of NAGS, and the evolutionary relationship between the two NAGS groups.


Asunto(s)
N-Acetiltransferasa de Aminoácidos/química , Secuencia de Aminoácidos , N-Acetiltransferasa de Aminoácidos/metabolismo , Animales , Bacterias/enzimología , Dominio Catalítico , Humanos , Datos de Secuencia Molecular
8.
Mol Genet Metab ; 113(1-2): 127-30, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25135652

RESUMEN

The Urea Cycle Disorders Consortium (UCDC) is a member of the NIH funded Rare Diseases Clinical Research Network and is performing a longitudinal study of 8 urea cycle disorders (UCDs) with initial enrollment beginning in 2006. The consortium consists of 14 sites in the U.S., Canada and Europe. This report summarizes data mining studies of 614 patients with UCDs enrolled in the UCDC's longitudinal study protocol. The most common disorder is ornithine transcarbamylase deficiency, accounting for more than half of the participants. We calculated the overall prevalence of urea cycle disorders to be 1/35,000, with 2/3rds presenting initial symptoms after the newborn period. We found the mortality rate to be 24% in neonatal onset cases and 11% in late onset cases. The most common precipitant of clinical hyperammonemic episodes in the post-neonatal period was intercurrent infections. Elevations in both blood ammonia and glutamine appeared to be biomarkers for neurocognitive outcome. In terms of chronic treatment, low protein diet appeared to result in normal weight but decreased linear growth while N-scavenger therapy with phenylbutyrate resulted in low levels of branched chain amino acids. Finally, we found an unexpectedly high risk for hepatic dysfunction in patients with ornithine transcarbamylase deficiency. This natural history study illustrates how a collaborative study of a rare genetic disorder can result in an improved understanding of morbidity and disease outcome.


Asunto(s)
Trastornos Innatos del Ciclo de la Urea/diagnóstico , Trastornos Innatos del Ciclo de la Urea/terapia , Edad de Inicio , Biomarcadores/metabolismo , Humanos , Estudios Longitudinales , Morbilidad , Mortalidad , Prevalencia , Resultado del Tratamiento , Trastornos Innatos del Ciclo de la Urea/epidemiología
9.
J Pediatr ; 165(2): 401-403.e3, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24880889

RESUMEN

Identical studies using stable isotopes were performed before and after a 3-day trial of oral N-carbamyl-l-glutamate (NCG) in 5 subjects with late-onset carbamyl phosphate synthetase deficiency. NCG augmented ureagenesis and decreased plasma ammonia in 4 of 5 subjects. There was marked improvement in nitrogen metabolism with long-term NCG administration in 1 subject.


Asunto(s)
Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/tratamiento farmacológico , Glutamatos/uso terapéutico , Glutamina/sangre , Urea/metabolismo , Adolescente , Adulto , Amoníaco/sangre , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/sangre , Niño , Preescolar , Femenino , Humanos , Modelos Lineales , Masculino , Espectrometría de Masas , Resultado del Tratamiento , Adulto Joven
10.
Proteins ; 81(10): 1847-54, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23609986

RESUMEN

We report herein the crystal structure of Escherichia coli RimK at a resolution of 2.85 Å, an enzyme that catalyzes the post-translational addition of up to 15 C-terminal glutamate residues to ribosomal protein S6. The structure belongs to the ATP-grasp superfamily and is organized as a tetramer, consistent with gel filtration analysis. Each subunit consists of three distinct structural domains and the active site is located in the cleft between these domains. The catalytic reaction appears to occur at the junction between the three domains as ATP binds between the B and C domains, and other substrates bind nearby.


Asunto(s)
Proteínas de Escherichia coli , Péptido Sintasas , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Sitios de Unión , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Péptido Sintasas/química , Péptido Sintasas/metabolismo , Conformación Proteica , Pliegue de Proteína
11.
Biochem Biophys Res Commun ; 430(4): 1253-8, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23261468

RESUMEN

N-Acetyl-L-glutamate synthase catalyzes the conversion of AcCoA and glutamate to CoA and N-acetyl-L-glutamate (NAG), the first step of the arginine biosynthetic pathway in lower organisms. In mammals, NAG is an obligate cofactor of carbamoyl phosphate synthetase I in the urea cycle. We have previously reported the structures of NAGS from Neisseria gonorrhoeae (ngNAGS) with various substrates bound. Here we reported the preparation of the bisubstrate analog, CoA-S-acetyl-L-glutamate, the crystal structure of ngNAGS with CoA-NAG bound, and kinetic studies of several active site mutants. The results are consistent with a one-step nucleophilic addition-elimination mechanism with Glu353 as the catalytic base and Ser392 as the catalytic acid. The structure of the ngNAGS-bisubstrate complex together with the previous ngNAGS structures delineates the catalytic reaction path for ngNAGS.


Asunto(s)
Acilcoenzima A/química , N-Acetiltransferasa de Aminoácidos/química , Proteínas Bacterianas/química , Glutamatos/química , Neisseria gonorrhoeae/enzimología , N-Acetiltransferasa de Aminoácidos/genética , Proteínas Bacterianas/genética , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Estructura Secundaria de Proteína , Especificidad por Sustrato
12.
Biochem Biophys Res Commun ; 437(4): 585-90, 2013 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-23850694

RESUMEN

Maricaulis maris N-acetylglutamate synthase/kinase (mmNAGS/K) catalyzes the first two steps in L-arginine biosynthesis and has a high degree of sequence and structural homology to human N-acetylglutamate synthase, a regulator of the urea cycle. The synthase activity of both mmNAGS/K and human NAGS are regulated by L-arginine, although L-arginine is an allosteric inhibitor of mmNAGS/K, but an activator of human NAGS. To investigate the mechanism of allosteric inhibition of mmNAGS/K by L-arginine, we have determined the structure of the mmNAGS/K complexed with L-arginine at 2.8 Å resolution. In contrast to the structure of mmNAGS/K in the absence of L-arginine where there are conformational differences between the four subunits in the asymmetric unit, all four subunits in the L-arginine liganded structure have very similar conformations. In this conformation, the AcCoA binding site in the N-acetyltransferase (NAT) domain is blocked by a loop from the amino acid kinase (AAK) domain, as a result of a domain rotation that occurs when L-arginine binds. This structural change provides an explanation for the allosteric inhibition of mmNAGS/K and related enzymes by L-arginine. The allosterically regulated mechanism for mmNAGS/K differs significantly from that for Neisseria gonorrhoeae NAGS (ngNAGS). To define the active site, several residues near the putative active site were mutated and their activities determined. These experiments identify roles for Lys356, Arg386, Asn391 and Tyr397 in the catalytic mechanism.


Asunto(s)
Alphaproteobacteria/enzimología , N-Acetiltransferasa de Aminoácidos/química , Arginina/química , Proteínas Bacterianas/química , Sitio Alostérico , Catálisis , Dominio Catalítico , Escherichia coli/metabolismo , Mutagénesis , Mutagénesis Sitio-Dirigida , Mutación , Unión Proteica
13.
J Pediatr ; 162(2): 324-9.e1, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22901741

RESUMEN

OBJECTIVE: To compare the clinical course and outcome of patients diagnosed with one of 4 neonatal-onset urea cycle disorders (UCDs): deficiency of carbamyl phosphate synthase 1 (CPSD), ornithine transcarbamylase (OTCD), argininosuccinate synthase (ASD), or argininosuccinate lyase (ALD). STUDY DESIGN: Clinical, biochemical, and neuropsychological data from 103 subjects with neonatal-onset UCDs were derived from the Longitudinal Study of Urea Cycle Disorders, an observational protocol of the Urea Cycle Disorders Consortium, one of the Rare Disease Clinical Research Networks. RESULTS: Some 88% of the subjects presented clinically by age 7 days. Peak ammonia level was 963 µM in patients with proximal UCDs (CPSD or OTCD), compared with 589 µM in ASD and 573 µM in ALD. Roughly 25% of subjects with CPSD or OTCD, 18% of those with ASD, and 67% of those with ALD had a "honeymoon period," defined as the time interval from discharge from initial admission to subsequent admission for hyperammonemia, greater than 1 year. The proportion of patients with a poor outcome (IQ/Developmental Quotient <70) was greatest in ALD (68%), followed by ASD (54%) and CPSD/OTCD (47%). This trend was not significant, but was observed in both patients aged <4 years and those aged ≥ 4 years. Poor cognitive outcome was not correlated with peak ammonia level or duration of initial admission. CONCLUSION: Neurocognitive outcomes do not differ between patients with proximal UCDs and those with distal UCDs. Factors other than hyperammonemia may contribute to poor neurocognitive outcome in the distal UCDs.


Asunto(s)
Trastornos Innatos del Ciclo de la Urea/diagnóstico , Preescolar , Ensayos Clínicos como Asunto , Femenino , Humanos , Lactante , Recién Nacido , Estudios Longitudinales , Masculino , Trastornos Innatos del Ciclo de la Urea/complicaciones
14.
J Biol Chem ; 286(25): 22055-68, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21540182

RESUMEN

We previously reported that isobutylmethylxanthine (IBMX), a derivative of oxypurine, inhibits citrulline synthesis by an as yet unknown mechanism. Here, we demonstrate that IBMX and other oxypurines containing a 2,6-dione group interfere with the binding of glutamate to the active site of N-acetylglutamate synthetase (NAGS), thereby decreasing synthesis of N-acetylglutamate, the obligatory activator of carbamoyl phosphate synthase-1 (CPS1). The result is reduction of citrulline and urea synthesis. Experiments were performed with (15)N-labeled substrates, purified hepatic CPS1, and recombinant mouse NAGS as well as isolated mitochondria. We also used isolated hepatocytes to examine the action of various oxypurines on ureagenesis and to assess the ameliorating affect of N-carbamylglutamate and/or l-arginine on NAGS inhibition. Among various oxypurines tested, only IBMX, xanthine, or uric acid significantly increased the apparent K(m) for glutamate and decreased velocity of NAGS, with little effect on CPS1. The inhibition of NAGS is time- and dose-dependent and leads to decreased formation of the CPS1-N-acetylglutamate complex and consequent inhibition of citrulline and urea synthesis. However, such inhibition was reversed by supplementation with N-carbamylglutamate. The data demonstrate that xanthine and uric acid, both physiologically occurring oxypurines, inhibit the hepatic synthesis of N-acetylglutamate. An important and novel concept emerging from this study is that xanthine and/or uric acid may have a role in the regulation of ureagenesis and, thus, nitrogen homeostasis in normal and disease states.


Asunto(s)
N-Acetiltransferasa de Aminoácidos/antagonistas & inhibidores , Regulación hacia Abajo/efectos de los fármacos , Hígado/metabolismo , Urea/metabolismo , Ácido Úrico/farmacología , Xantina/farmacología , 1-Metil-3-Isobutilxantina/farmacología , N-Acetiltransferasa de Aminoácidos/aislamiento & purificación , N-Acetiltransferasa de Aminoácidos/metabolismo , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/aislamiento & purificación , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Citrulina/biosíntesis , Relación Dosis-Respuesta a Droga , Glutamatos/biosíntesis , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Cinética , Hígado/citología , Hígado/enzimología , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Proteins ; 80(5): 1436-47, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22328207

RESUMEN

Putrescine carbamoyltransferase (PTCase) catalyzes the conversion of carbamoylputrescine to putrescine and carbamoyl phosphate (CP), a substrate of carbamate kinase (CK). The crystal structure of PTCase has been determined and refined at 3.2 Å resolution. The trimeric molecular structure of PTCase is similar to other carbamoyltransferases, including the catalytic subunit of aspartate carbamoyltransferase (ATCase) and ornithine carbamoyltransferase (OTCase). However, in contrast to other trimeric carbamoyltransferases, PTCase binds both CP and putrescine with Hill coefficients at saturating concentrations of the other substrate of 1.53 ± 0.03 and 1.80 ± 0.06, respectively. PTCase also has a unique structural feature: a long C-terminal helix that interacts with the adjacent subunit to enhance intersubunit interactions in the molecular trimer. The C-terminal helix appears to be essential for both formation of the functional trimer and catalytic activity, since truncated PTCase without the C-terminal helix aggregates and has only 3% of native catalytic activity. The active sites of PTCase and OTCase are similar, with the exception of the 240's loop. PTCase lacks the proline-rich sequence found in knotted carbamoyltransferases and is unknotted. A Blast search of all available genomes indicates that 35 bacteria, most of which are Gram-positive, have an agcB gene encoding PTCase located near the genes that encode agmatine deiminase and CK, consistent with the catabolic role of PTCase in the agmatine degradation pathway. Sequence comparisons indicate that the C-terminal helix identified in this PTCase structure will be found in all other PTCases identified, suggesting that it is the signature feature of the PTCase family of enzymes.


Asunto(s)
Proteínas Bacterianas/química , Transferasas de Carboxilo y Carbamoilo/química , Enterococcus faecalis/enzimología , Regulación Alostérica , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Transferasas de Carboxilo y Carbamoilo/metabolismo , Dominio Catalítico , Cristalización , Bases de Datos Genéticas , Enterococcus faecalis/metabolismo , Histidina , Datos de Secuencia Molecular , Ornitina Carbamoiltransferasa , Conformación Proteica , Subunidades de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia
16.
Mol Genet Metab ; 106(2): 160-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22503289

RESUMEN

All knockout mouse models of urea cycle disorders die in the neonatal period or shortly thereafter. Since N-acetylglutamate synthase (NAGS) deficiency in humans can be effectively treated with N-carbamyl-l-glutamate (NCG), we sought to develop a mouse model of this disorder that could be rescued by biochemical intervention, reared to adulthood, reproduce, and become a novel animal model for hyperammonemia. Founder NAGS knockout heterozygous mice were obtained from the trans-NIH Knock-Out Mouse Project. Genotyping of the mice was performed by PCR and confirmed by Western blotting of liver and intestine. NCG and L-citrulline (Cit) were used to rescue the NAGS knockout homozygous (Nags(-/-)) pups and the rescued animals were characterized. We observed an 85% survival rate of Nags(-/-) mice when they were given intraperitoneal injections with NCG and Cit during the newborn period until weaning and supplemented subsequently with both compounds in their drinking water. This regimen has allowed for normal development, apparent health, and reproduction. Interruption of this rescue intervention resulted in the development of severe hyperammonemia and death within 48 h. In addition to hyperammonemia, interruption of rescue supplementation was associated with elevated plasma glutamine, glutamate, and lysine, and reduced citrulline, arginine, ornithine and proline levels. We conclude that NAGS deprived mouse model has been developed which can be rescued by NCG and Cit and reared to reproduction and beyond. This biochemically salvageable mouse model recapitulates the clinical phenotype of proximal urea cycle disorders and can be used as a reliable model of induced hyperammonemia by manipulating the administration of the rescue compounds.


Asunto(s)
N-Acetiltransferasa de Aminoácidos/deficiencia , Modelos Animales de Enfermedad , Hiperamonemia/enzimología , Ratones , N-Acetiltransferasa de Aminoácidos/genética , N-Acetiltransferasa de Aminoácidos/metabolismo , Animales , Cruzamiento , Femenino , Orden Génico , Marcación de Gen , Genotipo , Glutamatos/uso terapéutico , Humanos , Hiperamonemia/tratamiento farmacológico , Hiperamonemia/genética , Hiperamonemia/mortalidad , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
17.
Hum Mutat ; 32(10): 1153-60, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21681857

RESUMEN

N-acetylglutamate synthase (NAGS) catalyzes the conversion of glutamate and acetyl-CoA to NAG, the essential allosteric activator of carbamyl phosphate synthetase I, the first urea cycle enzyme in mammals. A 17-year-old female with recurrent hyperammonemia attacks, the cause of which remained undiagnosed for 8 years in spite of multiple molecular and biochemical investigations, showed markedly enhanced ureagenesis (measured by isotope incorporation) in response to N-carbamylglutamate (NCG). This led to sequencing of the regulatory regions of the NAGS gene and identification of a deleterious single-base substitution in the upstream enhancer. The homozygous mutation (c.-3064C>A), affecting a highly conserved nucleotide within the hepatic nuclear factor 1 (HNF-1) binding site, was not found in single nucleotide polymorphism databases and in a screen of 1,086 alleles from a diverse population. Functional assays demonstrated that this mutation decreases transcription and binding of HNF-1 to the NAGS gene, while a consensus HNF-1 binding sequence enhances binding to HNF-1 and increases transcription. Oral daily NCG therapy restored ureagenesis in this patient, normalizing her biochemical markers, and allowing discontinuation of alternate pathway therapy and normalization of her diet with no recurrence of hyperammonemia. Inc.


Asunto(s)
N-Acetiltransferasa de Aminoácidos/genética , Elementos de Facilitación Genéticos , Glutamatos/uso terapéutico , Eliminación de Secuencia , Trastornos Innatos del Ciclo de la Urea/tratamiento farmacológico , Trastornos Innatos del Ciclo de la Urea/genética , Adolescente , Alelos , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Niño , Femenino , Frecuencia de los Genes , Glutamatos/metabolismo , Células Hep G2 , Factor Nuclear 1 del Hepatocito/metabolismo , Humanos , Motivos de Nucleótidos , Polimorfismo de Nucleótido Simple , Alineación de Secuencia , Resultado del Tratamiento , Trastornos Innatos del Ciclo de la Urea/metabolismo
18.
Biochemistry ; 49(32): 6887-95, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20695527

RESUMEN

N-Acetyl-l-ornithine transcarbamylase (AOTCase), rather than ornithine transcarbamylase (OTCase), is the essential carbamylase enzyme in the arginine biosynthesis of several plant and human pathogens. The specificity of this unique enzyme provides a potential target for controlling the spread of these pathogens. Recently, several crystal structures of AOTCase from Xanthomonas campestris (xc) have been determined. In these structures, an unexplained electron density at the tip of the Lys302 side chain was observed. Using (13)C NMR spectroscopy, we show herein that Lys302 is post-translationally carboxylated. The structure of wild-type AOTCase in a complex with the bisubstrate analogue N(delta)-(phosphonoacetyl)-N(alpha)-acetyl-l-ornithine (PALAO) indicates that the carboxyl group on Lys302 forms a strong hydrogen bonding network with surrounding active site residues, Lys252, Ser253, His293, and Glu92 from the adjacent subunit either directly or via a water molecule. Furthermore, the carboxyl group is involved in binding N-acetyl-l-ornithine via a water molecule. Activity assays with the wild-type enzyme and several mutants demonstrate that the post-translational modification of lysine 302 has an important role in catalysis.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Enlace de Hidrógeno , Lisina/metabolismo , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Ornitina/análogos & derivados , Ornitina/metabolismo , Ornitina Carbamoiltransferasa/química , Ornitina Carbamoiltransferasa/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Estructura Secundaria de Proteína , Xanthomonas campestris/enzimología
19.
Mol Genet Metab ; 100 Suppl 1: S13-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20303810

RESUMEN

N-acetylglutamate (NAG) is a unique enzyme cofactor, essential for liver ureagenesis in mammals while it is the first committed substrate for de novo arginine biosynthesis in microorganisms and plants. The enzyme that produces NAG from glutamate and CoA, NAG synthase (NAGS), is allosterically inhibited by arginine in microorganisms and plants and activated in mammals. This transition of the allosteric effect occurred when tetrapods moved from sea to land. The first mammalian NAGS gene (from mouse) was cloned in 2002 and revealed significant differences from the NAGS ortholog in microorganisms. Almost all NAGS genes possess a C-terminus transferase domain in which the catalytic activity resides and an N-terminus kinase domain where arginine binds. The three-dimensional structure of NAGS shows two distinctly folded domains. The kinase domain binds arginine while the acetyltransferase domain contains the catalytic site. NAGS deficiency in humans leads to hyperammonemia and can be primary, due to mutations in the NAGS gene or secondary due to other mitochondrial aberrations that interfere with the normal function of the same enzyme. For either condition, N-carbamylglutamate (NCG), a stable functional analog of NAG, was found to either restore or improve the deficient urea-cycle function.


Asunto(s)
N-Acetiltransferasa de Aminoácidos/química , N-Acetiltransferasa de Aminoácidos/metabolismo , Secuencia de Aminoácidos , N-Acetiltransferasa de Aminoácidos/deficiencia , N-Acetiltransferasa de Aminoácidos/genética , Animales , Biocatálisis , Evolución Molecular , Humanos , Datos de Secuencia Molecular , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Trastornos Innatos del Ciclo de la Urea/enzimología , Trastornos Innatos del Ciclo de la Urea/terapia
20.
Mol Genet Metab ; 100 Suppl 1: S37-41, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20338795

RESUMEN

Stable isotopes have been an invaluable adjunct to biomedical research for more than 70years. Indeed, the isotopic approach has revolutionized our understanding of metabolism, revealing it to be an intensely dynamic process characterized by an unending cycle of synthesis and degradation. Isotopic studies have taught us that the urea cycle is intrinsic to such dynamism, since it affords a capacious mechanism by which to eliminate waste nitrogen when rates of protein degradation (or dietary protein intake) are especially high. Isotopes have enabled an appreciation of the degree to which ureagenesis is compromised in patients with urea cycle defects. Indeed, isotopic studies of urea cycle flux correlate well with the severity of cognitive impairment in these patients. Finally, the use of isotopes affords an ideal tool with which to gauge the efficacy of therapeutic interventions to augment residual flux through the cycle.


Asunto(s)
Marcaje Isotópico/métodos , Urea/metabolismo , N-Acetiltransferasa de Aminoácidos/deficiencia , N-Acetiltransferasa de Aminoácidos/metabolismo , Cloruro de Amonio/administración & dosificación , Cloruro de Amonio/farmacología , Dióxido de Carbono/metabolismo , Isótopos de Carbono/metabolismo , Humanos , Urea/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA