Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Orphanet J Rare Dis ; 19(1): 168, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38637895

RESUMO

BACKGROUND: The autosomal recessive disorder N-acetylglutamate synthase (NAGS) deficiency is the rarest defect of the urea cycle, with an incidence of less than one in 2,000,000 live births. Hyperammonemic crises can be avoided in individuals with NAGS deficiency by the administration of carbamylglutamate (also known as carglumic acid), which activates carbamoyl phosphatase synthetase 1 (CPS1). The aim of this case series was to introduce additional cases of NAGS deficiency to the literature as well as to assess the role of nutrition management in conjunction with carbamylglutamate therapy across new and existing cases. METHODS: We conducted retrospective chart reviews of seven cases of NAGS deficiency in the US and Canada, focusing on presentation, diagnosis, medication management, nutrition management, and outcomes. RESULTS: Five new and two previously published cases were included. Presenting symptoms were consistent with previous reports. Diagnostic confirmation via molecular testing varied in protocol across cases, with consecutive single gene tests leading to long delays in diagnosis in some cases. All patients responded well to carbamylglutamate therapy, as indicated by normalization of plasma ammonia and citrulline, as well as urine orotic acid in patients with abnormal levels at baseline. Although protein restriction was not prescribed in any cases after carbamylglutamate initiation, two patients continued to self-restrict protein intake. One patient experienced two episodes of hyperammonemia that resulted in poor long-term outcomes. Both episodes occurred after a disruption in access to carbamylglutamate, once due to insurance prior authorization requirements and language barriers and once due to seizure activity limiting the family's ability to administer carbamylglutamate. CONCLUSIONS: Follow-up of patients with NAGS deficiency should include plans for illness and for disruption of carbamylglutamate access, including nutrition management strategies such as protein restriction. Carbamylglutamate can help patients with NAGS deficiency to liberalize their diets, but the maximum safe level of protein intake to prevent hyperammonemia is not yet known. Patients using this medication should still monitor their diet closely and be prepared for any disruptions in medication access, which might require immediate dietary adjustments or medical intervention to prevent hyperammonemia.


Assuntos
Glutamatos , Hiperamonemia , Distúrbios Congênitos do Ciclo da Ureia , Humanos , Aminoácido N-Acetiltransferase/genética , Aminoácido N-Acetiltransferase/metabolismo , Hiperamonemia/tratamento farmacológico , Estudos Retrospectivos
2.
Microb Cell Fact ; 22(1): 138, 2023 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-37495979

RESUMO

BACKGROUND: L-arginine is an important amino acid with applications in diverse industrial and pharmaceutical fields. N-acetylglutamate, synthesized from L-glutamate and acetyl-CoA, is a precursor of the L-arginine biosynthetic branch in microorganisms. The enzyme that produces N-acetylglutamate, N-acetylglutamate synthase, is allosterically inhibited by L-arginine. L-glutamate, as a central metabolite, provides carbon backbone for diverse biological compounds besides L-arginine. When glucose is the sole carbon source, the theoretical maximum carbon yield towards L-arginine is 96.7%, but the experimental highest yield was 51%. The gap of L-arginine yield indicates the regulation complexity of carbon flux and energy during the L-arginine biosynthesis. Besides endogenous biosynthesis, N-acetylglutamate, the key precursor of L-arginine, can be obtained by chemical acylation of L-glutamate with a high yield of 98%. To achieve high-yield production of L-arginine, we demonstrated a novel approach by directly feeding precursor N-acetylglutamate to engineered Escherichia coli. RESULTS: We reported a new approach for the high yield of L-arginine production in E. coli. Gene argA encoding N-acetylglutamate synthase was deleted to disable endogenous biosynthesis of N-acetylglutamate. The feasibility of external N-acetylglutamate towards L-arginine was verified via growth assay in argA- strain. To improve L-arginine production, astA encoding arginine N-succinyltransferase, speF encoding ornithine decarboxylase, speB encoding agmatinase, and argR encoding an arginine responsive repressor protein were disrupted. Based on overexpression of argDGI, argCBH operons, encoding enzymes of the L-arginine biosynthetic pathway, ~ 4 g/L L-arginine was produced in shake flask fermentation, resulting in a yield of 0.99 mol L-arginine/mol N-acetylglutamate. This strain was further engineered for the co-production of L-arginine and pyruvate by removing genes adhE, ldhA, poxB, pflB, and aceE, encoding enzymes involved in the conversion and degradation of pyruvate. The resulting strain was shown to produce 4 g/L L-arginine and 11.3 g/L pyruvate in shake flask fermentation. CONCLUSIONS: Here, we developed a novel approach to avoid the strict regulation of L-arginine on ArgA and overcome the metabolism complexity in the L-arginine biosynthesis pathway. We achieve a high yield of L-arginine production from N-acetylglutamate in E. coli. Co-production pyruvate and L-arginine was used as an example to increase the utilization of input carbon sources.


Assuntos
Escherichia coli , Ácido Glutâmico , Escherichia coli/metabolismo , Aminoácido N-Acetiltransferase/metabolismo , Ácido Glutâmico/metabolismo , Arginina , Piruvatos/metabolismo , Carbono/metabolismo , Engenharia Metabólica/métodos
3.
Sci Rep ; 11(1): 3580, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33574402

RESUMO

The urea cycle protects the central nervous system from ammonia toxicity by converting ammonia to urea. N-acetylglutamate synthase (NAGS) catalyzes formation of N-acetylglutamate, an essential allosteric activator of carbamylphosphate synthetase 1. Enzymatic activity of mammalian NAGS doubles in the presence of L-arginine, but the physiological significance of NAGS activation by L-arginine has been unknown. The NAGS knockout (Nags-/-) mouse is an animal model of inducible hyperammonemia, which develops hyperammonemia without N-carbamylglutamate and L-citrulline supplementation (NCG + Cit). We used adeno associated virus (AAV) based gene transfer to correct NAGS deficiency in the Nags-/- mice, established the dose of the vector needed to rescue Nags-/- mice from hyperammonemia and measured expression levels of Nags mRNA and NAGS protein in the livers of rescued animals. This methodology was used to investigate the effect of L-arginine on ureagenesis in vivo by treating Nags-/- mice with AAV vectors encoding either wild-type or E354A mutant mouse NAGS (mNAGS), which is not activated by L-arginine. The Nags-/- mice expressing E354A mNAGS were viable but had elevated plasma ammonia concentration despite similar levels of the E354A and wild-type mNAGS proteins. The corresponding mutation in human NAGS (NP_694551.1:p.E360D) that abolishes binding and activation by L-arginine was identified in a patient with NAGS deficiency. Our results show that NAGS deficiency can be rescued by gene therapy, and suggest that L-arginine binding to the NAGS enzyme is essential for normal ureagenesis.


Assuntos
Aminoácido N-Acetiltransferase/genética , Técnicas de Transferência de Genes , Hiperamonemia/genética , Distúrbios Congênitos do Ciclo da Ureia/genética , Aminoácido N-Acetiltransferase/metabolismo , Animais , Arginina/metabolismo , Arginina/farmacologia , Citrulina/metabolismo , Citrulina/farmacologia , Dependovirus/genética , Modelos Animais de Doenças , Glutamatos/metabolismo , Glutamatos/farmacologia , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Hiperamonemia/terapia , Camundongos , Camundongos Knockout , Proteínas Mutantes/genética , Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/patologia , Distúrbios Congênitos do Ciclo da Ureia/terapia
4.
Biochimie ; 183: 89-99, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33309754

RESUMO

Despite biochemical and genetic testing being the golden standards for identification of proximal urea cycle disorders (UCDs), genotype-phenotype correlations are often unclear. Co-occurring partial defects affecting more than one gene have not been demonstrated so far in proximal UCDs. Here, we analyzed the mutational spectrum of 557 suspected proximal UCD individuals. We probed oligomerizing forms of NAGS, CPS1 and OTC, and evaluated the surface exposure of residues mutated in heterozygously affected individuals. BN-PAGE and gel-filtration chromatography were employed to discover protein-protein interactions within recombinant enzymes. From a total of 281 confirmed patients, only 15 were identified as "heterozygous-only" candidates (i.e. single defective allele). Within these cases, the only missense variants to potentially qualify as dominant negative triggers were CPS1 p.Gly401Arg and NAGS p.Thr181Ala and p.Tyr512Cys, as assessed by residue oligomerization capacity and surface exposure. However, all three candidates seem to participate in critical intramolecular functions, thus, unlikely to facilitate protein-protein interactions. This interpretation is further supported by BN-PAGE and gel-filtration analyses revealing no multiprotein proximal urea cycle complex formation. Collectively, genetic analysis, structural considerations and in vitro experiments point against a prominent role of dominant negative effects in human proximal UCDs.


Assuntos
Aminoácido N-Acetiltransferase , Carbamoil-Fosfato Sintase (Amônia) , Genes Dominantes , Mutação de Sentido Incorreto , Ornitina Carbamoiltransferase , Distúrbios Congênitos do Ciclo da Ureia , Substituição de Aminoácidos , Aminoácido N-Acetiltransferase/química , Aminoácido N-Acetiltransferase/genética , Aminoácido N-Acetiltransferase/metabolismo , Carbamoil-Fosfato Sintase (Amônia)/química , Carbamoil-Fosfato Sintase (Amônia)/genética , Carbamoil-Fosfato Sintase (Amônia)/metabolismo , Feminino , Heterozigoto , Homozigoto , Humanos , Masculino , Ornitina Carbamoiltransferase/química , Ornitina Carbamoiltransferase/genética , Ornitina Carbamoiltransferase/metabolismo , Domínios Proteicos , Distúrbios Congênitos do Ciclo da Ureia/enzimologia , Distúrbios Congênitos do Ciclo da Ureia/genética
5.
Dokl Biochem Biophys ; 495(1): 334-337, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33368046

RESUMO

Three-dimensional full-atom model of the enzyme complex with acetyl-CoA and substrate was constructed on the basis of the primary sequence of amino acid residues of N-acetyl glutamate synthase. Bioinformatics approaches of computer modeling were applied, including multiple sequence alignment, prediction of co-evolutionary contacts, and ab initio folding. On the basis of the results of calculations by classical molecular dynamics and combined quantum and molecular mechanics (QM/MM) methods, the structure of the active site and the reaction mechanism of N-acetylglutamate formation are described. Agreement of the structures of the enzyme-product complexes obtained in computer modeling and in the X-ray studies validates the reliability of modeling predictions.


Assuntos
Aminoácido N-Acetiltransferase/química , Aminoácido N-Acetiltransferase/metabolismo , Neisseria gonorrhoeae/enzimologia , Catálise , Domínio Catalítico , Simulação por Computador , Cristalografia por Raios X , Modelos Moleculares , Neisseria gonorrhoeae/química , Neisseria gonorrhoeae/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
6.
Am J Physiol Gastrointest Liver Physiol ; 318(5): G912-G927, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32174131

RESUMO

Glucagon regulates the hepatic amino acid metabolism and increases ureagenesis. Ureagenesis is activated by N-acetylglutamate (NAG), formed via activation of N-acetylglutamate synthase (NAGS). With the aim to identify the steps whereby glucagon both acutely and chronically regulates ureagenesis, we investigated whether glucagon receptor-mediated activation of ureagenesis is required in a situation where NAGS activity and/or NAG levels are sufficient to activate the first step of the urea cycle in vivo. Female C57BL/6JRj mice treated with a glucagon receptor antagonist (GRA), glucagon receptor knockout (Gcgr-/-) mice, and wild-type (Gcgr+/+) littermates received an intraperitoneal injection of N-carbamoyl glutamate (Car; a stable variant of NAG), l-citrulline (Cit), Car and Cit (Car + Cit), or PBS. In separate experiments, Gcgr-/- and Gcgr+/+ mice were administered N-carbamoyl glutamate and l-citrulline (wCar + wCit) in the drinking water for 8 wk. Car, Cit, and Car + Cit significantly (P < 0.05) increased plasma urea concentrations, independently of pharmacological and genetic disruption of glucagon receptor signaling (P = 0.9). Car increased blood glucose concentrations equally in GRA- and vehicle-treated mice (P = 0.9), whereas the increase upon Car + Cit was impaired in GRA-treated mice (P = 0.008). Blood glucose concentrations remained unchanged in Gcgr-/- mice upon Car (P = 0.2) and Car + Cit (P = 0.9). Eight weeks administration of wCar + wCit did not change blood glucose (P > 0.2), plasma amino acid (P > 0.4), and urea concentrations (P > 0.3) or the area of glucagon-positive cells (P > 0.3) in Gcgr-/- and Gcgr+/+ mice. Our data suggest that glucagon-mediated activation of ureagenesis is not required when NAGS activity and/or NAG levels are sufficient to activate the first step of the urea cycle.NEW & NOTEWORTHY Hepatic ureagenesis is essential in amino acid metabolism and is importantly regulated by glucagon, but the exact mechanism is unclear. With the aim to identify the steps whereby glucagon both acutely and chronically regulates ureagenesis, we here show, contrary to our hypothesis, that glucagon receptor-mediated activation of ureagenesis is not required when N-acetylglutamate synthase activity and/or N-acetylglutamate levels are sufficient to activate the first step of the urea cycle in vivo.


Assuntos
Citrulina/administração & dosagem , Glucagon/metabolismo , Glutamatos/administração & dosagem , Fígado/efeitos dos fármacos , Receptores de Glucagon/deficiência , Receptores de Glucagon/metabolismo , Ureia/sangue , Aminoácido N-Acetiltransferase/metabolismo , Animais , Carbamoil-Fosfato Sintase (Amônia)/metabolismo , Feminino , Glutamatos/metabolismo , Antagonistas de Hormônios/administração & dosagem , Fígado/enzimologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Glucagon/antagonistas & inibidores , Receptores de Glucagon/genética
7.
Sci Rep ; 8(1): 15436, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30337552

RESUMO

N-acetylglutamate synthase deficiency (NAGSD, MIM #237310) is an autosomal recessive disorder of the urea cycle that results from absent or decreased production of N-acetylglutamate (NAG) due to either decreased NAGS gene expression or defective NAGS enzyme. NAG is essential for the activity of carbamylphosphate synthetase 1 (CPS1), the first and rate-limiting enzyme of the urea cycle. NAGSD is the only urea cycle disorder that can be treated with a single drug, N-carbamylglutamate (NCG), which can activate CPS1 and completely restore ureagenesis in patients with NAGSD. We describe a novel sequence variant NM_153006.2:c.-3026C > T in the NAGS enhancer that was found in three patients from two families with NAGSD; two patients had hyperammonemia that resolved upon treatment with NCG, while the third patient increased dietary protein intake after initiation of NCG therapy. Two patients were homozygous for the variant while the third patient had the c.-3026C > T variant and a partial uniparental disomy that encompassed the NAGS gene on chromosome 17. The c.-3026C > T sequence variant affects a base pair that is highly conserved in vertebrates; the variant is predicted to be deleterious by several bioinformatics tools. Functional assays in cultured HepG2 cells demonstrated that the c.-3026C > T substitution could result in reduced expression of the NAGS gene. These findings underscore the importance of analyzing NAGS gene regulatory regions when looking for molecular causes of NAGSD.


Assuntos
Aminoácido N-Acetiltransferase/genética , Elementos Facilitadores Genéticos , Variação Genética , Distúrbios Congênitos do Ciclo da Ureia/etiologia , Aminoácido N-Acetiltransferase/metabolismo , Sequência de Bases , Criança , Pré-Escolar , Feminino , Humanos , Hiperamonemia , Prognóstico , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/patologia
8.
Int J Mol Sci ; 19(2)2018 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-29364180

RESUMO

N-acetylglutamate synthase deficiency (NAGSD) is an extremely rare urea cycle disorder (UCD) with few adult cases so far described. Diagnosis of late-onset presentations is difficult and delayed treatment may increase the risk of severe hyperammonemia. We describe a 52-year-old woman with recurrent headaches who experienced an acute onset of NAGSD. As very few papers focus on headaches in UCDs, we also report a literature review of types and pathophysiologic mechanisms of UCD-related headaches. In our case, headaches had been present since puberty (3-4 days a week) and were often accompanied by nausea, vomiting, or behavioural changes. Despite three previous episodes of altered consciousness, ammonia was measured for the first time at 52 years and levels were increased. Identification of the new homozygous c.344C>T (p.Ala115Val) NAGS variant allowed the definite diagnosis of NAGSD. Bioinformatic analysis suggested that an order/disorder alteration of the mutated form could affect the arginine-binding site, resulting in poor enzyme activation and late-onset presentation. After optimized treatment for NAGSD, ammonia and amino acid levels were constantly normal and prevented other headache bouts. The manuscript underlies that headache may be the presenting symptom of UCDs and provides clues for the rapid diagnosis and treatment of late-onset NAGSD.


Assuntos
Distúrbios Congênitos do Ciclo da Ureia/diagnóstico , Idade de Início , Aminoácido N-Acetiltransferase/metabolismo , Biomarcadores , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Eletroencefalografia , Feminino , Glutamatos/uso terapêutico , Humanos , Pessoa de Meia-Idade , Avaliação de Sintomas , Resultado do Tratamento , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/patologia , Distúrbios Congênitos do Ciclo da Ureia/terapia
9.
Nat Chem Biol ; 13(6): 640-646, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28369041

RESUMO

Toxin-antitoxin (TA) loci are prevalent in bacterial genomes. They are suggested to play a central role in dormancy and persister states. Under normal growth conditions, TA toxins are neutralized by their cognate antitoxins, and under stress conditions, toxins are freed and inhibit essential cellular processes using a variety of mechanisms. Here we characterize ataR-ataT, a novel TA system, from enterohemorrhagic Escherichia coli. We show that the toxin AtaT is a GNAT family enzyme that transfers an acetyl group from acetyl coenzyme A to the amine group of the methionyl aminoacyl moiety of initiator tRNA. AtaT specifically modifies Met-tRNAfMet, but no other aminoacyl-tRNAs, including the elongator Met-tRNAMet. We demonstrate that once acetylated, AcMet-tRNAfMet fails to interact with initiation factor-2 (IF2), resulting in disruption of the translation initiation complex. This work reveals a new mechanism of translation inhibition and confirms Met-tRNAfMet as a prime target to efficiently block cell growth.


Assuntos
Aminoácido N-Acetiltransferase/metabolismo , Escherichia coli , Regulação da Expressão Gênica/genética , RNA de Transferência de Metionina/metabolismo , Acetilação , Eletroforese em Gel Bidimensional , Modelos Biológicos , Biossíntese de Proteínas
10.
Biochemistry ; 56(6): 805-808, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28135072

RESUMO

The biosynthesis of the azabicyclic ring system of the azinomycin family of antitumor agents represents the "crown jewel" of the pathway and is a complex process involving at least 14 enzymatic steps. This study reports on the first biosynthetic step, the inroads, in the construction of the novel aziridino [1,2-a]pyrrolidine, azabicyclic core, allowing us to support a new mechanism for azabicycle formation.


Assuntos
Aldeído Oxirredutases/metabolismo , Aminoácido N-Acetiltransferase/metabolismo , Antineoplásicos Alquilantes/metabolismo , Compostos Azabicíclicos/metabolismo , Proteínas de Bactérias/metabolismo , Desenho de Fármacos , Fosfotransferases (Aceptor do Grupo Carboxila)/metabolismo , Pirrolidinas/metabolismo , Acetilcoenzima A/metabolismo , Acetilação , Aldeído Oxirredutases/genética , Aminoácido N-Acetiltransferase/genética , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacologia , Compostos Azabicíclicos/química , Compostos Azabicíclicos/farmacologia , Proteínas de Bactérias/genética , Biocatálise , Dipeptídeos/química , Dipeptídeos/metabolismo , Dipeptídeos/farmacologia , Técnicas de Inativação de Genes , Ácido Glutâmico/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Estrutura Molecular , Mutação , Naftalenos/química , Naftalenos/metabolismo , Naftalenos/farmacologia , Peptídeos/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Fosfotransferases (Aceptor do Grupo Carboxila)/genética , Pirrolidinas/química , Pirrolidinas/farmacologia , Proteínas Recombinantes/metabolismo , Streptomyces/enzimologia , Streptomyces/metabolismo , Especificidade por Substrato
11.
Sci Rep ; 6: 38711, 2016 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-27934952

RESUMO

N-acetylglutamate synthase (NAGS; E.C.2.3.1.1) catalyzes the formation of N-acetylglutamate (NAG) from acetyl coenzyme A and glutamate. In microorganisms and plants, NAG is the first intermediate of the L-arginine biosynthesis; in animals, NAG is an allosteric activator of carbamylphosphate synthetase I and III. In some bacteria bifunctional N-acetylglutamate synthase-kinase (NAGS-K) catalyzes the first two steps of L-arginine biosynthesis. L-arginine inhibits NAGS in bacteria, fungi, and plants and activates NAGS in mammals. L-arginine increased thermal stability of the NAGS-K from Maricaulis maris (MmNAGS-K) while it destabilized the NAGS-K from Xanthomonas campestris (XcNAGS-K). Analytical gel chromatography and ultracentrifugation indicated tetrameric structure of the MmMNAGS-K in the presence and absence of L-arginine and a tetramer-octamer equilibrium that shifted towards tetramers upon binding of L-arginine for the XcNAGS-K. Analytical gel chromatography of mouse NAGS (mNAGS) indicated either different oligomerization states that are in moderate to slow exchange with each other or deviation from the spherical shape of the mNAGS protein. The partition coefficient of the mNAGS increased in the presence of L-arginine suggesting smaller hydrodynamic radius due to change in either conformation or oligomerization. Different effects of L-arginine on oligomerization of NAGS may have implications for efforts to determine the three-dimensional structure of mammalian NAGS.


Assuntos
Alphaproteobacteria/enzimologia , Aminoácido N-Acetiltransferase/química , Arginina/química , Proteínas de Bactérias/química , Multimerização Proteica , Xanthomonas campestris/enzimologia , Aminoácido N-Acetiltransferase/metabolismo , Animais , Arginina/metabolismo , Proteínas de Bactérias/metabolismo , Estrutura Quaternária de Proteína
12.
Mol Genet Metab ; 119(4): 307-310, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27771289

RESUMO

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.


Assuntos
Aminoácido N-Acetiltransferase/genética , Carbamoil-Fosfato Sintase (Amônia)/genética , Hiperamonemia/diagnóstico , Distúrbios Congênitos do Ciclo da Ureia/diagnóstico , Acetilcoenzima A/metabolismo , Aminoácido N-Acetiltransferase/metabolismo , Animais , Carbamoil-Fosfato Sintase (Amônia)/deficiência , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatologia , Fígado/enzimologia , Camundongos , Camundongos Knockout , Espectrometria de Massas em Tandem , Distúrbios Congênitos do Ciclo da Ureia/genética , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/fisiopatologia
13.
Hum Mutat ; 37(7): 679-94, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27037498

RESUMO

N-acetyl-L-glutamate synthase (NAGS) deficiency (NAGSD), the rarest urea cycle defect, is clinically indistinguishable from carbamoyl phosphate synthetase 1 deficiency, rendering the identification of NAGS gene mutations key for differentiation, which is crucial, as only NAGSD has substitutive therapy. Over the last 13 years, we have identified 43 patients from 33 families with NAGS mutations, of which 14 were novel. Overall, 36 NAGS mutations have been found so far in 56 patients from 42 families, of which 76% are homozygous for the mutant allele. 61% of mutations are missense changes. Lack or decrease of NAGS protein is predicted for ∼1/3 of mutations. Missense mutations frequency is inhomogeneous along NAGS: null for exon 1, but six in exon 6, which reflects the paramount substrate binding/catalytic role of the C-terminal domain (GNAT domain). Correspondingly, phenotypes associated with missense mutations mapping in the GNAT domain are more severe than phenotypes of amino acid kinase domain-mapping missense mutations. Enzyme activity and stability assays with 12 mutations introduced into pure recombinant Pseudomonas aeruginosa NAGS, together with in silico structural analysis, support the pathogenic role of most NAGSD-associated mutations found. The disease-causing mechanisms appear to be, from higher to lower frequency, decreased solubility/stability, aberrant kinetics/catalysis, and altered arginine modulation.


Assuntos
Aminoácido N-Acetiltransferase/genética , Mutação de Sentido Incorreto , Distúrbios Congênitos do Ciclo da Ureia/genética , Aminoácido N-Acetiltransferase/química , Aminoácido N-Acetiltransferase/metabolismo , Predisposição Genética para Doença , Humanos , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Estabilidade Proteica
14.
Small GTPases ; 6(4): 189-95, 2015 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-26507377

RESUMO

The small GTP-binding protein Ran is involved in the regulation of essential cellular processes in interphase but also in mitotic cells: Ran controls the nucleocytoplasmic transport of proteins and RNA, it regulates mitotic spindle formation and nuclear envelope assembly. Deregulations in Ran dependent processes were implicated in the development of severe diseases such as cancer and neurodegenerative disorders. To understand how Ran-function is regulated is therefore of highest importance. Recently, several lysine-acetylation sites in Ran were identified by quantitative mass-spectrometry, some being located in highly important regions such as the P-loop, switch I, switch II and the G5/SAK motif. We recently reported that lysine-acetylation regulates nearly all aspects of Ran-function such as RCC1 catalyzed nucleotide exchange, intrinsic nucleotide hydrolysis, its interaction with NTF2 and the formation of import- and export-complexes. As a hint for its biological importance, we identified Ran-specific lysine-deacetylases (KDACs) and -acetyltransferases (KATs). Also for other small GTPases such as Ras, Rho, Cdc42, and for many effectors and regulators thereof, lysine-acetylation sites were discovered. However, the functional impact of lysine-acetylation as a regulator of protein function has only been marginally investigated so far. We will discuss recent findings of lysine-acetylation as a novel modification to regulate Ras-protein signaling.


Assuntos
Transdução de Sinais , Proteína ran de Ligação ao GTP/metabolismo , Proteínas ras/metabolismo , Acetilação , Motivos de Aminoácidos , Aminoácido N-Acetiltransferase/genética , Aminoácido N-Acetiltransferase/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Lisina/genética , Lisina/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína ran de Ligação ao GTP/genética , Proteínas ras/genética
15.
Sci Rep ; 5: 15029, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26461067

RESUMO

Folate metabolism is central to cell proliferation and a target of commonly used cancer chemotherapeutics. In particular, the mitochondrial folate-coupled metabolism is thought to be important for proliferating cancer cells. The enzyme MTHFD2 in this pathway is highly expressed in human tumors and broadly required for survival of cancer cells. Although the enzymatic activity of the MTHFD2 protein is well understood, little is known about its larger role in cancer cell biology. We here report that MTHFD2 is co-expressed with two distinct gene sets, representing amino acid metabolism and cell proliferation, respectively. Consistent with a role for MTHFD2 in cell proliferation, MTHFD2 expression was repressed in cells rendered quiescent by deprivation of growth signals (serum) and rapidly re-induced by serum stimulation. Overexpression of MTHFD2 alone was sufficient to promote cell proliferation independent of its dehydrogenase activity, even during growth restriction. In addition to its known mitochondrial localization, we found MTHFD2 to have a nuclear localization and co-localize with DNA replication sites. These findings suggest a previously unknown role for MTHFD2 in cancer cell proliferation, adding to its known function in mitochondrial folate metabolism.


Assuntos
Aminoácido N-Acetiltransferase/metabolismo , Núcleo Celular/enzimologia , Ácido Fólico/metabolismo , Metilenotetra-Hidrofolato Desidrogenase (NADP)/metabolismo , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Células HeLa , Humanos , Camundongos , Mitocôndrias/metabolismo , Proteínas Nucleares/metabolismo , Ratos , Especificidade da Espécie
16.
Virology ; 485: 58-78, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26210075

RESUMO

BACKGROUND: Borna disease virus (BDV) is a neurotropic RNA virus persistently infecting mammalian hosts including humans. Lysine acetylation (Kac) is a key protein post-translational modification (PTM). The unexpectedly broad regulatory scope of Kac let us to profile the entire acetylome upon BDV infection. METHODS: The acetylome was profiled through stable isotope labeling for cell culture (SILAC)-based quantitative proteomics. The quantifiable proteome was annotated using bioinformatics. RESULTS: We identified and quantified 791 Kac sites in 473 Kac proteins in human BDV Hu-H1-infected and non-infected oligodendroglial (OL) cells. Bioinformatic analysis revealed that BDV infection alters the acetylation of metabolic proteins, membrane-associated proteins and transmembrane transporter activity, and affects the acetylation of several lysine acetyltransferases (KAT). CONCLUSIONS: Upon BDV persistence the OL acetylome is manipulated towards higher energy and transporter levels necessary for shuttling BDV proteins to and from nuclear replication sites.


Assuntos
Aminoácido N-Acetiltransferase/metabolismo , Vírus da Doença de Borna/fisiologia , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Oligodendroglia/metabolismo , Processamento de Proteína Pós-Traducional , Proteoma/metabolismo , Acetilação , Sequência de Aminoácidos , Aminoácido N-Acetiltransferase/genética , Proteínas de Transporte/genética , Linhagem Celular , Biologia Computacional , Metabolismo Energético , Feto , Interações Hospedeiro-Patógeno , Humanos , Marcação por Isótopo , Lisina/metabolismo , Proteínas de Membrana/genética , Anotação de Sequência Molecular , Dados de Sequência Molecular , Oligodendroglia/virologia , Mapeamento de Interação de Proteínas , Proteoma/genética , Replicação Viral
17.
Chem Biol ; 22(8): 1030-1039, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26190825

RESUMO

The finding that chromatin modifications are sensitive to changes in cellular cofactor levels potentially links altered tumor cell metabolism and gene expression. However, the specific enzymes and metabolites that connect these two processes remain obscure. Characterizing these metabolic-epigenetic axes is critical to understanding how metabolism supports signaling in cancer, and developing therapeutic strategies to disrupt this process. Here, we describe a chemical approach to define the metabolic regulation of lysine acetyltransferase (KAT) enzymes. Using a novel chemoproteomic probe, we identify a previously unreported interaction between palmitoyl coenzyme A (palmitoyl-CoA) and KAT enzymes. Further analysis reveals that palmitoyl-CoA is a potent inhibitor of KAT activity and that fatty acyl-CoA precursors reduce cellular histone acetylation levels. These studies implicate fatty acyl-CoAs as endogenous regulators of histone acetylation, and suggest novel strategies for the investigation and metabolic modulation of epigenetic signaling.


Assuntos
Acil Coenzima A/metabolismo , Aminoácido N-Acetiltransferase/metabolismo , Histona Acetiltransferases/metabolismo , Lisina/metabolismo , Acetilação , Acil Coenzima A/biossíntese , Acil Coenzima A/química , Aminoácido N-Acetiltransferase/química , Células HEK293 , Histona Acetiltransferases/química , Humanos , Cinética , Lisina/química , Modelos Químicos , Palmitoil Coenzima A/química , Palmitoil Coenzima A/metabolismo , Proteômica
18.
Int J Mol Sci ; 16(6): 13004-22, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26068232

RESUMO

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.


Assuntos
Aminoácido N-Acetiltransferase/química , Sequência de Aminoácidos , Aminoácido N-Acetiltransferase/metabolismo , Animais , Bactérias/enzimologia , Domínio Catalítico , Humanos , Dados de Sequência Molecular
19.
J Proteomics ; 114: 214-25, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25464366

RESUMO

Even though protein initiator methionine excision (NME) and N-terminal acetylation (NTA) have been relatively well investigated in eukaryotic proteomes, few studies were dedicated to these modifications in bacteria up to now. In this work, we investigated, for the first time, the N-terminal proteome of the bacterium Pseudomonas aeruginosa PA14 by studying the NME and NTA processes using proteomic approaches. For NME, most of proteins had their initiator Met cleaved (63%) and the nature of the penultimate residue seems to be essential for this cleavage. Concerning NTA, two methods were applied (protein fractionation and peptide enrichment). This allowed us to identify 117 Nα-acetylated proteins, among them 113 have not yet been described as modified in bacteria. Most often, the non-acetylated form was over-represented compared to the acetylated form, arguing that this latter was a minor part of the total abundance of a given protein. Furthermore, some proteins with acetylated initiator methionine were observed. The present work significantly enlarges the number of N-terminally modified proteins in bacteria and confirms that these modifications are a general and fundamental process, not only restricted to eukaryotes. BIOLOGICAL SIGNIFICANCE: Protein modifications in prokaryotes have been detected more recently than in eukaryotes. Methionine cleavage and N-terminal acetylation are two common protein N-terminal modifications. Despite their importance in bacterial processes, they are less investigated. The characterization of N-terminal acetylation in bacteria is a challenge because no antibody exists and it is a less frequent modification than in eukaryotes. We used proteomic approaches (enrichment, fractionation, nanoLC-MS/MS, and bioinformatic analyses) to investigate the N-terminal methionine excision and to profile the N-terminal acetylome of P. aeruginosa strain PA14. From our results, around 60% of the proteins had their iMet cleaved. In total, 117 proteins were identified constituting the largest dataset in prokaryotes. Among them, proteins kept their initiator methionine and were acetylated. These results may facilitate the design of experiments to better understand the role of acetylation at the protein N-terminus of P. aeruginosa PA14.


Assuntos
Proteínas de Bactérias/metabolismo , Processamento de Proteína Pós-Traducional , Pseudomonas aeruginosa/metabolismo , Acetilação , Sequência de Aminoácidos , Aminoácido N-Acetiltransferase/metabolismo , Proteínas de Bactérias/análise , Metionina/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/metabolismo , Proteoma/análise , Proteoma/metabolismo , Proteômica
20.
PLoS One ; 9(1): e85597, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465614

RESUMO

The urea cycle converts ammonia, a waste product of protein catabolism, into urea. Because fish dispose ammonia directly into water, the role of the urea cycle in fish remains unknown. Six enzymes, N-acetylglutamate synthase (NAGS), carbamylphosphate synthetase III, ornithine transcarbamylase, argininosuccinate synthase, argininosuccinate lyase and arginase 1, and two membrane transporters, ornithine transporter and aralar, comprise the urea cycle. The genes for all six enzymes and both transporters are present in the zebrafish genome. NAGS (EC 2.3.1.1) catalyzes the formation of N-acetylglutamate from glutamate and acetyl coenzyme A and in zebrafish is partially inhibited by L-arginine. NAGS and other urea cycle genes are highly expressed during the first four days of zebrafish development. Sequence alignment of NAGS proteins from six fish species revealed three regions of sequence conservation: the mitochondrial targeting signal (MTS) at the N-terminus, followed by the variable and conserved segments. Removal of the MTS yields mature zebrafish NAGS (zfNAGS-M) while removal of the variable segment from zfNAGS-M results in conserved NAGS (zfNAGS-C). Both zfNAGS-M and zfNAGS-C are tetramers in the absence of L-arginine; addition of L-arginine decreased partition coefficients of both proteins. The zfNAGS-C unfolds over a broader temperature range and has higher specific activity than zfNAGS-M. In the presence of L-arginine the apparent Vmax of zfNAGS-M and zfNAGS-C decreased, their Km(app) for acetyl coenzyme A increased while the Km(app) for glutamate remained unchanged. The expression pattern of NAGS and other urea cycle genes in developing zebrafish suggests that they may have a role in citrulline and/or arginine biosynthesis during the first day of development and in ammonia detoxification thereafter. Biophysical and biochemical properties of zebrafish NAGS suggest that the variable segment may stabilize a tetrameric state of zfNAGS-M and that under physiological conditions zebrafish NAGS catalyzes formation of N-acetylglutamate at the maximal rate.


Assuntos
Aminoácido N-Acetiltransferase/genética , Embrião não Mamífero/metabolismo , Perfilação da Expressão Gênica , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Acetilcoenzima A/metabolismo , Sequência de Aminoácidos , Aminoácido N-Acetiltransferase/química , Aminoácido N-Acetiltransferase/metabolismo , Animais , Arginina/farmacologia , Biocatálise/efeitos dos fármacos , Embrião não Mamífero/embriologia , Embrião não Mamífero/enzimologia , Estabilidade Enzimática , Regulação da Expressão Gênica no Desenvolvimento , Glutamatos/metabolismo , Ácido Glutâmico/metabolismo , Cinética , Dados de Sequência Molecular , Peso Molecular , Multimerização Proteica , Desdobramento de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Temperatura , Fatores de Tempo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA