Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
J Inherit Metab Dis ; 47(2): 220-229, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38375550

ABSTRACT

Carbamoyl phosphate synthetase 1 (CPS1) and ornithine transcarbamylase (OTC) deficiencies are rare urea cycle disorders, which can lead to life-threatening hyperammonemia. Liver transplantation (LT) provides a cure and offers an alternative to medical treatment and life-long dietary restrictions with permanent impending risk of hyperammonemia. Nevertheless, in most patients, metabolic aberrations persist after LT, especially low plasma citrulline levels, with questionable clinical impact. So far, little is known about these alterations and there is no consensus, whether l-citrulline substitution after LT improves patients' symptoms and outcomes. In this multicentre, retrospective, observational study of 24 patients who underwent LT for CPS1 (n = 11) or OTC (n = 13) deficiency, 25% did not receive l-citrulline or arginine substitution. Correlation analysis revealed no correlation between substitution dosage and citrulline levels (CPS1, p = 0.8 and OTC, p = 1). Arginine levels after liver transplantation were normal after LT independent of citrulline substitution. Native liver survival had no impact on mental impairment (p = 0.67). Regression analysis showed no correlation between l-citrulline substitution and failure to thrive (p = 0.611) or neurological outcome (p = 0.701). Peak ammonia had a significant effect on mental impairment (p = 0.017). Peak plasma ammonia levels correlate with mental impairment after LT in CPS1 and OTC deficiency. Growth and intellectual impairment after LT are not significantly associated with l-citrulline substitution.


Subject(s)
Hyperammonemia , Liver Transplantation , Ornithine Carbamoyltransferase Deficiency Disease , Humans , Ornithine Carbamoyltransferase Deficiency Disease/surgery , Hyperammonemia/drug therapy , Citrulline , Carbamyl Phosphate/metabolism , Carbamyl Phosphate/therapeutic use , Ammonia/metabolism , Retrospective Studies , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Arginine/therapeutic use , Ornithine Carbamoyltransferase
2.
Am J Physiol Gastrointest Liver Physiol ; 325(4): G334-G346, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37489865

ABSTRACT

Carbamoyl phosphate synthetase 1 (CPS1) is the most abundant hepatocyte mitochondrial matrix protein. Hypoosmotic stress increases CPS1 release in isolated mouse hepatocytes without cell death. We hypothesized that increased CPS1 release during hypoosmosis is selective and associates with altered mitochondrial morphology. Both ex vivo and in vivo models were assessed. Mouse hepatocytes and livers were challenged with isotonic or hypoosmotic (35 mosM) buffer. Mice were injected intraperitoneally with water (10% body weight) with or without an antidiuretic. Mitochondrial and cytosolic fractions were isolated using differential centrifugation, then analyzed by immunoblotting to assess subcellular redistribution of four mitochondrial proteins: CPS1, ornithine transcarbamylase (OTC), pyrroline-5-carboxylate reductase 1 (PYCR1), and cytochrome c. Mitochondrial morphology alterations were examined using electron microscopy. Hypoosmotic treatment of whole livers or hepatocytes led to preferential or increased mitochondrial release, respectively, of CPS1 as compared with two mitochondrial matrix proteins (OTC/PYCR1) and with the intermembrane space protein, cytochrome c. Mitochondrial apoptosis-induced channel opening using staurosporine in hepatocytes led to preferential CPS1 and cytochrome c release. The CPS1-selective changes were accompanied by dramatic alterations in ultrastructural mitochondrial morphology. In mice, hypoosmosis/hyponatremia led to increased liver vascular congestion and increased CPS1 in bile but not blood, coupled with mitochondrial structural alterations. In contrast, isotonic increase of intravascular volume led to a decrease in mitochondrial size with limited change in bile CPS1 compared with hypoosmotic conditions and absence of the hypoosmosis-associated histological alterations. Taken together, hepatocyte CPS1 is selectively released in response to hypoosmosis/hyponatremia and provides a unique biomarker of mitochondrial injury.NEW & NOTEWORTHY Exposure of isolated mouse livers, primary cultured hepatocytes, or mice to hypoosmosis/hyponatremia conditions induces significant mitochondrial shape alterations accompanied by preferential release of the mitochondrial matrix protein CPS1, a urea cycle enzyme. In contrast, the intermembrane space protein, cytochrome c, and two other matrix proteins, including the urea cycle enzyme ornithine transcarbamylase, remain preferentially retained in mitochondria. Therefore, hepatocyte CPS1 manifests unique mitochondrial stress response compartmentalization and is a sensitive sensor of mitochondrial hypoosmotic/hyponatremic injury.


Subject(s)
Hyponatremia , Liver Diseases , Animals , Mice , Carbamyl Phosphate/metabolism , Ornithine Carbamoyltransferase/metabolism , Cytochromes c/metabolism , Hyponatremia/metabolism , Hyponatremia/pathology , Hepatocytes/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Liver Diseases/metabolism , Mitochondria/metabolism , Urea/metabolism
3.
Bioorg Chem ; 130: 106253, 2023 01.
Article in English | MEDLINE | ID: mdl-36356370

ABSTRACT

CPS1, the rate-limiting enzyme that controls the first reaction of the urea cycle, is responsible for converting toxic ammonia into non-toxic urea in mammals. While disruption of the functions of CPS1 leads to elevated ammonia and nerve damage in the body, mainly manifested as urea cycle disorder. Moreover, accumulating evidence has recently revealed that CPS1 is involved in a variety of human diseases, including CPS1D, cardiovascular disease, cancers, and others. In particular, CPS1 expression varies among cancers, being overexpressed in some cancers and downregulated in others, suggesting that CPS1 may be a promising cancer therapeutic target. In addition, some small-molecule inhibitors of CPS1 have been reported, which have not been confirmed experimentally in malignancies, meaning their future role is far from certain. In this review, we describe the structure and function of CPS1, highlight its important roles in various human diseases, and further discuss the potential diagnostic and therapeutic implications of small molecule compounds targeting CPS1.


Subject(s)
Carbamoyl-Phosphate Synthase I Deficiency Disease , Animals , Humans , Carbamoyl-Phosphate Synthase I Deficiency Disease/pathology , Carbamoyl-Phosphate Synthase I Deficiency Disease/therapy , Carbamyl Phosphate/metabolism , Ammonia/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/chemistry , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Urea , Mammals/metabolism
4.
Plant Biol (Stuttg) ; 25(1): 131-141, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36178874

ABSTRACT

Carbamoyl phosphate synthetase (CPS) catalyses the synthesis of ammonia carbamoyl phosphate (CP), which plays a key role in the biosynthesis of arginine and pyrimidine nucleotides. There are two subunits of the CPS enzyme in Populus trichocarpa, CarA (small subunit) and CarB (large subunit). Only when they coexist can CPS catalyse synthesis of CP. However, it is not clear how CPS responds to nitrogen (N) to affect arginine and pyrimidine nucleotide biosynthesis. In this study, bioinformatics methods were used to analyse the expression patterns of genes encoding CarA and CarB, and qRT-PCR and RNA-seq were used to investigate their molecular responses under different N concentrations. Phylogenetic analysis revealed that the phylogenetic trees of CarA and CarB had similar topologies. qRT-PCR showed that the PtCarA and PtCarB genes were regulated by N, while their N-regulated patterns differed in different tissues. The expression patterns of PtCarA and PtCarB show a significant positive correlation according to qRT-PCR and RNA-seq. The analysis of promoter cis-acting elements showed that the promoter regions of PtCarA1, PtCarA2 and PtCarB contained some identical cis-acting elements. According to analysis of the phylogenetic tree, expression patterns and promoter elements, we speculate that there might be coevolution among PtCarA1, PtCarA2 and PtCarB. This study provides valuable information for further understanding the function of CPS in poplar, especially for N response, and provides new ideas for studying the evolution of gene families related to heteromultimers.


Subject(s)
Populus , Populus/genetics , Populus/metabolism , Carbamyl Phosphate/metabolism , Phylogeny , Nitrogen , Arginine/genetics
5.
J Microbiol Biotechnol ; 32(12): 1527-1536, 2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36384810

ABSTRACT

Escherichia coli can use allantoin as its sole nitrogen source under anaerobic conditions. The ureidoglycolate produced by double release of ammonia from allantoin can flow into either the glyoxylate shunt or further catabolic transcarbamoylation. Although the former pathway is well studied, the genes of the latter (catabolic) pathway are not known. In the catabolic pathway, ureidoglycolate is finally converted to carbamoyl phosphate (CP) and oxamate, and then CP is dephosphorylated to carbamate by a catabolic carbamate kinase (CK), whereby ATP is formed. We identified the ybcF gene in a gene cluster containing fdrA-ylbE-ylbF-ybcF that is located downstream of the allDCE-operon. Reverse transcription PCR of total mRNA confirmed that the genes fdrA, ylbE, ylbF, and ybcF are co-transcribed. Deletion of ybcF caused only a slight increase in metabolic flow into the glyoxylate pathway, probably because CP was used to de novo synthesize pyrimidine and arginine. The activity of the catabolic CK was analyzed using purified YbcF protein. The Vmax is 1.82 U/mg YbcF for CP and 1.94 U/mg YbcF for ADP, and the KM value is 0.47 mM for CP and 0.43 mM for ADP. With these results, it was experimentally revealed that the ybcF gene of E. coli encodes catabolic CK, which completes anaerobic allantoin degradation through substrate-level phosphorylation. Therefore, we suggest renaming the ybcF gene as allK.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Allantoin , Carbamyl Phosphate/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Glyoxylates , Membrane Proteins , Phosphotransferases (Carboxyl Group Acceptor)/genetics , Phosphotransferases (Carboxyl Group Acceptor)/metabolism
6.
J Clin Lab Anal ; 36(10): e24692, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36098904

ABSTRACT

OBJECTIVE: The sensitivity and specificity of current biomarkers for gastric cancer were insufficient. The aim of the present study was to screen novel biomarkers and determine the diagnostic values of ornithine aminotransferase (OAT) and carbamoyl phosphate synthetase 1 (CPS1) for detecting gastric cancer. METHODS: With stable isotope tags, we labelled an initial discovery group of four paired gastric cancer tissue samples and identified with LC-ESI-MS/MS. A validation group of 159 gastric cancer samples and 30 healthy controls were used to validate the candidate targets. GSEA was used to explore the pathways activated in gastric cancer. RESULTS: Four hundred and thirty one proteins were found differentially expressed in gastric cancer tissues. Of these proteins, OAT and CPS1 were found over-expressed in gastric cancer patients, with sensitivity of 70.4% (95% CI: 63.3%-77.6%) and specificity of 80.5% (95% CI: 74.3%-86.7%) for ornithine aminotransferase, and with sensitivity of 68.6% (95% CI: 61.3%-75.8%) and specificity of 73% (95% CI: 66%-79.9%) for carbamoyl phosphate synthetase 1. The co-expression of OAT and CPS1 in gastric cancer tissues has a sensitivity of 81% (95% CI: 73.2%-88.8%) and specificity of 89% (95% CI: 83%-95%). Furthermore, both OAT and CPS1 were overexpressed in patients with local invasion T3 and T4 stages than those in patients with T1 and T2 stages. The co-expression of OAT and CPS1 was strongly correlated with histological grade I 68% (95% CI: 58.7%-77.3%) and TNM stage I/II 52% (95% CI: 42%-62%). The areas under ROC curves were up to 0.758 for the co-expression of OAT and CPS1 in gastric cancer. GSEA results showed that two gene sets and 30 gene sets were activated in OAT high- and CPS1 high-expression patients with gastric cancer, respectively. CONCLUSIONS: The present findings indicated a tight correlation between the co-expression of OAT and CPS1 and the histological grade, local invasion, and TNM stages of gastric cancer. Therefore, OAT and CPS1 might be predictors for gastric cancer invasion and potential targets for anticancer drug design for gastric cancer.


Subject(s)
Antineoplastic Agents , Stomach Neoplasms , Ammonia , Biomarkers , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carbamyl Phosphate/metabolism , Humans , Ornithine-Oxo-Acid Transaminase/genetics , Stomach Neoplasms/pathology , Tandem Mass Spectrometry
7.
Extremophiles ; 25(1): 15-24, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33084979

ABSTRACT

Here, we have analyzed the enzyme ornithine carbamoyltransferase (OCTase) in different classes of microorganisms belonging to psychrophiles, mesophiles and thermophiles. This OCTase catalyzes the formation of citrulline from carbamoyl phosphate (CP) and ornithine (ORN) in arginine biosynthesis pathway and has certain unique adaptations to regulate metabolic pathways in extreme conditions. The tertiary structure of OCTase showed two binding domains, the CP domain and ORN-binding domain at N and C terminals, respectively. We propose general acid-base catalysis in Pseudomonas gessardii between His259 and Asp220 in which later may act as a recipient of proton in the process. The comparative docking analysis showed that substrate-binding loops have been evolved to accommodate their lifestyles across the physiological temperature range where two substrates bind on two distinct loops in psychrophiles and mesophiles, whereas both the substrates bind on a single-substrate-binding loop in thermophiles and bring down the flexibility of the active site pocket to improve its evolutionary fitness.


Subject(s)
Carbamyl Phosphate/metabolism , Extremophiles/enzymology , Ornithine Carbamoyltransferase/chemistry , Pseudomonas/enzymology , Binding Sites , Catalysis , Molecular Docking Simulation , Ornithine Carbamoyltransferase/genetics , Protein Domains
8.
PLoS One ; 15(2): e0228487, 2020.
Article in English | MEDLINE | ID: mdl-32027716

ABSTRACT

Understanding how enzymes achieve their tremendous catalytic power is a major question in biochemistry. Greater understanding is also needed for enzyme engineering applications. In many cases, enzyme efficiency and specificity depend on residues not in direct contact with the substrate, termed remote residues. This work focuses on Escherichia coli ornithine transcarbamoylase (OTC), which plays a central role in amino acid metabolism. OTC has been reported to undergo an induced-fit conformational change upon binding its first substrate, carbamoyl phosphate (CP), and several residues important for activity have been identified. Using computational methods based on the computed chemical properties from theoretical titration curves, sequence-based scores derived from evolutionary history, and protein surface topology, residues important for catalytic activity were predicted. The roles of these residues in OTC activity were tested by constructing mutations at predicted positions, followed by steady-state kinetics assays and substrate binding studies with the variants. First-layer mutations R57A and D231A, second-layer mutation H272L, and third-layer mutation E299Q, result in 57- to 450-fold reductions in kcat/KM with respect to CP and 44- to 580-fold reductions with respect to ornithine. Second-layer mutations D140N and Y160S also reduce activity with respect to ornithine. Most variants had decreased stability relative to wild-type OTC, with variants H272L, H272N, and E299Q having the greatest decreases. Variants H272L, E299Q, and R57A also show compromised CP binding. In addition to direct effects on catalytic activity, effects on overall protein stability and substrate binding were observed that reveal the intricacies of how these residues contribute to catalysis.


Subject(s)
Escherichia coli/enzymology , Ornithine Carbamoyltransferase/chemistry , Ornithine Carbamoyltransferase/metabolism , Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Amino Acid Sequence , Amino Acid Substitution/genetics , Base Sequence , Binding Sites , Carbamyl Phosphate/chemistry , Carbamyl Phosphate/metabolism , Catalysis , Escherichia coli/genetics , Escherichia coli/metabolism , Kinetics , Mutagenesis, Site-Directed , Ornithine/metabolism , Ornithine Carbamoyltransferase/genetics , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs/genetics , Substrate Specificity/genetics
9.
Enzyme Microb Technol ; 129: 109354, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31307577

ABSTRACT

Carbamoyl phosphate synthetase (CPS) is a key enzyme in both pyrimidine and arginine biosynthesis. However, it is inhibited strongly by uridine monophosphate (UMP), which is an intermediate of the de-novo synthesis of pyrimidine nucleoside. In this study, the native carbamoyl phosphate synthetase, from Escherichia coli, was evolved by site-directed mutation and casting error-prone PCR. Compared with the wild-type, the variant N1015 F had released sensitivity to UMP and exhibited 100% of the initial activity in the presence of UMP. Variant K1006A exhibited 0.14-fold improvement in initial activity and kept above 65% of relative activity under the saturated concentration of inhibitor. Structure analysis of variants demonstrated that the reduced sensitivity to inhibitor was largely attributed to the decreased hydrogen bonds, which could reduce the binding affinity with UMP. Also, Phe with large side chain could narrow the binding pocket and generate more steric hindrance. Based on the results in this study, N1015F was an ideal alternative catalyst for the wild-type CPS for pyrimidine biosynthesis.


Subject(s)
Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli/enzymology , Carbamyl Phosphate/metabolism , Carbon-Nitrogen Ligases/metabolism , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Mutagenesis, Site-Directed , Mutation , Polymerase Chain Reaction , Protein Engineering , Pyrimidines/metabolism , Uridine Monophosphate/metabolism
10.
Hum Gene Ther ; 30(11): 1385-1394, 2019 11.
Article in English | MEDLINE | ID: mdl-31215258

ABSTRACT

Metabolic liver diseases are attractive gene therapy targets that necessitate reconstitution of enzymatic activity in functionally complex biochemical pathways. The levels of enzyme activity required in individual hepatocytes and the proportion of the hepatic cell mass that must be gene corrected for therapeutic benefit vary in a disease-dependent manner that is difficult to predict. While empirical evaluation is inevitably required, useful insights can nevertheless be gained from knowledge of disease pathophysiology and theoretical approaches such as mathematical modeling. Urea cycle defects provide an excellent example. Building on a previously described one-compartment model of the urea cycle, we have constructed a two-compartment model that can simulate liver-targeted gene therapy interventions using the computational program Mathematica. The model predicts that therapeutically effective reconstitution of ureagenesis will correlate most strongly with the proportion of the hepatic cell mass transduced rather than the level of enzyme-encoding transgene expression achieved in individual hepatocytes. Importantly, these predictions are supported by experimental data in mice and human genotype/phenotype correlations. The most notable example of the latter is ornithine transcarbamylase deficiency (X-linked) where impairment of ureagenesis in male and female patients is closely simulated by the one- and two-compartment models, respectively. Collectively, these observations support the practical value of mathematical modeling in evaluation of the disease-specific gene transfer challenges posed by complex metabolic phenotypes.


Subject(s)
Genetic Therapy , Models, Biological , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/therapy , Carbamyl Phosphate/metabolism , Computer Simulation , Humans , Ornithine Carbamoyltransferase Deficiency Disease/genetics
11.
Amino Acids ; 50(12): 1647-1661, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30238253

ABSTRACT

In all organisms, carbamoylphosphate (CP) is a precursor common to the synthesis of arginine and pyrimidines. In Escherichia coli and most other Gram-negative bacteria, CP is produced by a single enzyme, carbamoylphosphate synthase (CPSase), encoded by the carAB operon. This particular situation poses a question of basic physiological interest: what are the metabolic controls coordinating the synthesis and distribution of this high-energy substance in view of the needs of both pathways? The study of the mechanisms has revealed unexpected moonlighting gene regulatory activities of enzymes and functional links between mechanisms as diverse as gene regulation and site-specific DNA recombination. At the level of enzyme production, various regulatory mechanisms were found to cooperate in a particularly intricate transcriptional control of a pair of tandem promoters. Transcription initiation is modulated by an interplay of several allosteric DNA-binding transcription factors using effector molecules from three different pathways (arginine, pyrimidines, purines), nucleoid-associated factors (NAPs), trigger enzymes (enzymes with a second unlinked gene regulatory function), DNA remodeling (bending and wrapping), UTP-dependent reiterative transcription initiation, and stringent control by the alarmone ppGpp. At the enzyme level, CPSase activity is tightly controlled by allosteric effectors originating from different pathways: an inhibitor (UMP) and two activators (ornithine and IMP) that antagonize the inhibitory effect of UMP. Furthermore, it is worth noticing that all reaction intermediates in the production of CP are extremely reactive and unstable, and protected by tunneling through a 96 Å long internal channel.


Subject(s)
Carbamyl Phosphate/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Ligases/genetics , Arginine/biosynthesis , Escherichia coli/genetics , Promoter Regions, Genetic , Pyrimidines/biosynthesis
12.
Mol Genet Metab ; 124(4): 243-253, 2018 08.
Article in English | MEDLINE | ID: mdl-29801986

ABSTRACT

Carbamoyl phosphate synthetase 1 (CPS1) is a urea cycle enzyme that forms carbamoyl phosphate from bicarbonate, ammonia and ATP. Bi-allelic mutations of the CPS1 gene result in a urea cycle disorder presenting with hyperammonemia, often with reduced citrulline, and without orotic aciduria. CPS1 deficiency is particularly challenging to treat and lack of early recognition typically results in early neonatal death. Therapeutic interventions have limited efficacy and most patients develop long-term neurologic sequelae. Using transgenic techniques, we generated a conditional Cps1 knockout mouse. By loxP/Cre recombinase technology, deletion of the Cps1 locus was achieved in adult transgenic animals using a Cre recombinase-expressing adeno-associated viral vector. Within four weeks from vector injection, all animals developed hyperammonemia without orotic aciduria and died. Minimal CPS1 protein was detectable in livers. To investigate the efficacy of gene therapy for CPS deficiency following knock-down of hepatic endogenous CPS1 expression, we injected these mice with a helper-dependent adenoviral vector (HDAd) expressing the large murine CPS1 cDNA under control of the phosphoenolpyruvate carboxykinase promoter. Liver-directed HDAd-mediated gene therapy resulted in survival, normalization of plasma ammonia and glutamine, and 13% of normal Cps1 expression. A gender difference in survival suggests that female mice may require higher hepatic CPS1 expression. We conclude that this conditional murine model recapitulates the clinical and biochemical phenotype detected in human patients with CPS1 deficiency and will be useful to investigate ammonia-mediated neurotoxicity and for the development of cell- and gene-based therapeutic approaches.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/therapy , Genetic Therapy , Hyperammonemia/therapy , Ammonia/metabolism , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/therapeutic use , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/metabolism , Carbamoyl-Phosphate Synthase I Deficiency Disease/pathology , Carbamyl Phosphate/metabolism , Female , Gene Expression Regulation, Enzymologic , Glutamine/metabolism , Humans , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hyperammonemia/pathology , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Knockout , Mutation , Orotate Phosphoribosyltransferase/deficiency , Orotate Phosphoribosyltransferase/genetics , Orotidine-5'-Phosphate Decarboxylase/deficiency , Orotidine-5'-Phosphate Decarboxylase/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
13.
Appl Environ Microbiol ; 84(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29150502

ABSTRACT

Carbamate kinases catalyze the conversion of carbamate to carbamoyl phosphate, which is readily transformed into other compounds. Carbamate forms spontaneously from ammonia and carbon dioxide in aqueous solutions, so the kinases have potential for sequestrative utilization of the latter compounds. Here, we compare seven carbamate kinases from mesophilic, thermophilic, and hyperthermophilic sources. In addition to the known enzymes from Enterococcus faecalis and Pyrococcus furiosus, the previously unreported enzymes from the hyperthermophiles Thermococcus sibiricus and Thermococcus barophilus, the thermophiles Fervidobacterium nodosum and Thermosipho melanesiensis, and the mesophile Clostridium tetani were all expressed recombinantly, each in high yield. Only the clostridial enzyme did not show catalysis. In direct assays of carbamate kinase activity, the three hyperthermophilic enzymes display higher specific activities at elevated temperatures, greater stability, and remarkable substrate turnover at alkaline pH (9.9 to 11.4). Thermococcus barophilus and Thermococcus sibiricus carbamate kinases were found to be the most active when the enzymes were tested at 80°C, and maintained activity over broad temperature and pH ranges. These robust thermococcal enzymes therefore represent ideal candidates for biotechnological applications involving aqueous ammonia solutions, since nonbuffered 0.0001 to 1.0 M solutions have pH values of approximately 9.8 to 11.8. As proof of concept, here we also show that carbamoyl phosphate produced by the Thermococcus barophilus kinase is efficiently converted in situ to carbamoyl aspartate by aspartate transcarbamoylase from the same source organism. Using acetyl phosphate to simultaneously recycle the kinase cofactor ATP, at pH 9.9 carbamoyl aspartate is produced in high yield and directly from solutions of ammonia, carbon dioxide, and aspartate.IMPORTANCE Much of the nitrogen in animal wastes and used in fertilizers is commonly lost as ammonia in water runoff, from which it must be removed to prevent downstream pollution and evolution of nitrogenous greenhouse gases. Since carbamate kinases transform ammonia and carbon dioxide to carbamoyl phosphate via carbamate, and carbamoyl phosphate may be converted into other valuable compounds, the kinases provide a route for useful sequestration of ammonia, as well as of carbon dioxide, another greenhouse gas. At the same time, recycling the ammonia in chemical synthesis reduces the need for its energy-intensive production. However, robust catalysts are required for such biotransformations. Here we show that carbamate kinases from hyperthermophilic archaea display remarkable stability and high catalytic activity across broad ranges of pH and temperature, making them promising candidates for biotechnological applications. We also show that carbamoyl phosphate produced by the kinases may be efficiently used to produce carbamoyl aspartate.


Subject(s)
Alkalies/metabolism , Anabolic Agents/metabolism , Phosphotransferases (Carboxyl Group Acceptor)/metabolism , Temperature , Ammonia/metabolism , Carbamates/metabolism , Carbamyl Phosphate/metabolism , Catalysis , Clostridium tetani/enzymology , Clostridium tetani/genetics , Clostridium tetani/metabolism , Enterococcus faecalis/enzymology , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Enzyme Stability , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Protein Conformation , Pyrococcus furiosus/enzymology , Pyrococcus furiosus/genetics , Pyrococcus furiosus/metabolism , Thermococcus/enzymology , Thermococcus/genetics , Thermococcus/metabolism
14.
Amino Acids ; 49(9): 1543-1559, 2017 09.
Article in English | MEDLINE | ID: mdl-28741223

ABSTRACT

Citrulline was chemically isolated more than 100 years ago and is ubiquitous in animals, plants, bacteria, and fungi. Most of the research on plant citrulline metabolism and transport has been carried out in Arabidopsis thaliana and the Cucurbitaceae family, particularly in watermelon which accumulates this non-proteinogenic amino acid to very high levels. Industrially, citrulline is produced via specially optimized microbial strains; however, the amounts present in watermelon render it an economically viable source providing that other high-value compounds can be co-extracted. In this review, we provide an overview of our current understanding of citrulline biosynthesis, transport, and catabolism in plants additionally pointing out significant gaps in our knowledge which need to be closed by future experimentation. This includes the identification of further potential enzymes of citrulline metabolism as well as obtaining a far better spatial resolution of both sub-cellular and long-distance partitioning of citrulline. We further discuss what is known concerning the biological function of citrulline in plants paying particular attention to the proposed roles in scavenging of excess NH4+ and as a compatible solute.


Subject(s)
Arabidopsis/metabolism , Citrulline/biosynthesis , Cucurbitaceae/metabolism , Gene Expression Regulation, Plant , Ornithine Carbamoyltransferase/genetics , Adaptation, Physiological , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Ammonia/metabolism , Animals , Arabidopsis/genetics , Argininosuccinate Synthase/genetics , Argininosuccinate Synthase/metabolism , Bacteria/metabolism , Biological Transport , Carbamyl Phosphate/metabolism , Citrulline/genetics , Cucurbitaceae/genetics , Fungi/metabolism , Humans , Ornithine/metabolism , Ornithine Carbamoyltransferase/metabolism , Stress, Physiological
15.
Structure ; 25(6): 912-923.e5, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28552578

ABSTRACT

CAD, the multifunctional protein initiating and controlling de novo biosynthesis of pyrimidines in animals, self-assembles into ∼1.5 MDa hexamers. The structures of the dihydroorotase (DHO) and aspartate transcarbamoylase (ATC) domains of human CAD have been previously determined, but we lack information on how these domains associate and interact with the rest of CAD forming a multienzymatic unit. Here, we prove that a construct covering human DHO and ATC oligomerizes as a dimer of trimers and that this arrangement is conserved in CAD-like from fungi, which holds an inactive DHO-like domain. The crystal structures of the ATC trimer and DHO-like dimer from the fungus Chaetomium thermophilum confirm the similarity with the human CAD homologs. These results demonstrate that, despite being inactive, the fungal DHO-like domain has a conserved structural function. We propose a model that sets the DHO and ATC complex as the central element in the architecture of CAD.


Subject(s)
Aspartate Carbamoyltransferase/chemistry , Aspartate Carbamoyltransferase/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/chemistry , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Dihydroorotase/chemistry , Dihydroorotase/metabolism , Aspartate Carbamoyltransferase/genetics , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/genetics , Carbamyl Phosphate/chemistry , Carbamyl Phosphate/metabolism , Chaetomium/enzymology , Crystallography, X-Ray , Dihydroorotase/genetics , Humans , Microscopy, Electron , Models, Molecular , Mutagenesis, Site-Directed , Protein Domains , Protein Multimerization , Pyrimidines/biosynthesis
16.
Nature ; 546(7656): 168-172, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28538732

ABSTRACT

Metabolic reprogramming by oncogenic signals promotes cancer initiation and progression. The oncogene KRAS and tumour suppressor STK11, which encodes the kinase LKB1, regulate metabolism and are frequently mutated in non-small-cell lung cancer (NSCLC). Concurrent occurrence of oncogenic KRAS and loss of LKB1 (KL) in cells specifies aggressive oncological behaviour. Here we show that human KL cells and tumours share metabolomic signatures of perturbed nitrogen handling. KL cells express the urea cycle enzyme carbamoyl phosphate synthetase-1 (CPS1), which produces carbamoyl phosphate in the mitochondria from ammonia and bicarbonate, initiating nitrogen disposal. Transcription of CPS1 is suppressed by LKB1 through AMPK, and CPS1 expression correlates inversely with LKB1 in human NSCLC. Silencing CPS1 in KL cells induces cell death and reduces tumour growth. Notably, cell death results from pyrimidine depletion rather than ammonia toxicity, as CPS1 enables an unconventional pathway of nitrogen flow from ammonia into pyrimidines. CPS1 loss reduces the pyrimidine to purine ratio, compromises S-phase progression and induces DNA-polymerase stalling and DNA damage. Exogenous pyrimidines reverse DNA damage and rescue growth. The data indicate that the KL oncological genotype imposes a metabolic vulnerability related to a dependence on a cross-compartmental pathway of pyrimidine metabolism in an aggressive subset of NSCLC.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , DNA/biosynthesis , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Ammonia/metabolism , Animals , Bicarbonates/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamyl Phosphate/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Death , Cell Proliferation , DNA Damage/drug effects , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Female , Gene Silencing , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Male , Metabolomics , Mice , Mitochondria/metabolism , Nitrogen/metabolism , Protein Serine-Threonine Kinases/metabolism , Purines/metabolism , Pyrimidines/pharmacology , S Phase , Transcription, Genetic , Xenograft Model Antitumor Assays
17.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 7): 523-33, 2016 07.
Article in English | MEDLINE | ID: mdl-27380369

ABSTRACT

The de novo pyrimidine-biosynthesis pathway of Plasmodium falciparum is a promising target for antimalarial drug discovery. The parasite requires a supply of purines and pyrimidines for growth and proliferation and is unable to take up pyrimidines from the host. Direct (or indirect) inhibition of de novo pyrimidine biosynthesis via dihydroorotate dehydrogenase (PfDHODH), the fourth enzyme of the pathway, has already been shown to be lethal to the parasite. In the second step of the plasmodial pyrimidine-synthesis pathway, aspartate and carbamoyl phosphate are condensed to N-carbamoyl-L-aspartate and inorganic phosphate by aspartate transcarbamoylase (PfATC). In this paper, the 2.5 Šresolution crystal structure of PfATC is reported. The space group of the PfATC crystals was determined to be monoclinic P21, with unit-cell parameters a = 87.0, b = 103.8, c = 87.1 Å, α = 90.0, ß = 117.7, γ = 90.0°. The presented PfATC model shares a high degree of homology with the catalytic domain of Escherichia coli ATC. There is as yet no evidence of the existence of a regulatory domain in PfATC. Similarly to E. coli ATC, PfATC was modelled as a homotrimer in which each of the three active sites is formed at the oligomeric interface. Each active site comprises residues from two adjacent subunits in the trimer with a high degree of evolutional conservation. Here, the activity loss owing to mutagenesis of the key active-site residues is also described.


Subject(s)
Aspartate Carbamoyltransferase/chemistry , Aspartic Acid/chemistry , Carbamyl Phosphate/chemistry , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Amino Acid Sequence , Aspartate Carbamoyltransferase/genetics , Aspartate Carbamoyltransferase/metabolism , Aspartic Acid/metabolism , Binding Sites , Carbamyl Phosphate/metabolism , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Models, Molecular , Mutation , Plasmids/chemistry , Plasmids/metabolism , Plasmodium falciparum/enzymology , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
18.
Sci Rep ; 5: 16950, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26592762

ABSTRACT

Human carbamoyl phosphate synthetase (CPS1), a 1500-residue multidomain enzyme, catalyzes the first step of ammonia detoxification to urea requiring N-acetyl-L-glutamate (NAG) as essential activator to prevent ammonia/amino acids depletion. Here we present the crystal structures of CPS1 in the absence and in the presence of NAG, clarifying the on/off-switching of the urea cycle by NAG. By binding at the C-terminal domain of CPS1, NAG triggers long-range conformational changes affecting the two distant phosphorylation domains. These changes, concerted with the binding of nucleotides, result in a dramatic remodeling that stabilizes the catalytically competent conformation and the building of the ~35 Å-long tunnel that allows migration of the carbamate intermediate from its site of formation to the second phosphorylation site, where carbamoyl phosphate is produced. These structures allow rationalizing the effects of mutations found in patients with CPS1 deficiency (presenting hyperammonemia, mental retardation and even death), as exemplified here for some mutations.


Subject(s)
Ammonia/chemistry , Carbamoyl-Phosphate Synthase (Ammonia)/chemistry , Carbamyl Phosphate/chemistry , Glutamates/chemistry , Urea/chemistry , Amino Acid Motifs , Ammonia/metabolism , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carbamoyl-Phosphate Synthase I Deficiency Disease/enzymology , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/pathology , Carbamyl Phosphate/metabolism , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Glutamates/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Phosphorylation , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera , Substrate Specificity , Urea/metabolism
19.
J Bacteriol ; 197(20): 3294-306, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26260457

ABSTRACT

UNLABELLED: In two-component signal transduction, a sensor protein transmitter module controls cognate receiver domain phosphorylation. Most receiver domain sequences contain a small residue (Gly or Ala) at position T + 1 just distal to the essential Thr or Ser residue that forms part of the active site. However, some members of the NarL receiver subfamily have a large hydrophobic residue at position T + 1. Our laboratory previously isolated a NarL mutant in which the T + 1 residue Val-88 was replaced with an orthodox small Ala. This NarL V88A mutant confers a striking phenotype in which high-level target operon expression is both signal (nitrate) and sensor (NarX and NarQ) independent. This suggests that the NarL V88A protein is phosphorylated by cross talk from noncognate sources. Although cross talk was enhanced in ackA null strains that accumulate acetyl phosphate, it persisted in pta ackA double null strains that cannot synthesize this compound and was observed also in narL(+) strains. This indicates that acetate metabolism has complex roles in mediating NarL cross talk. Contrariwise, cross talk was sharply diminished in an arcB barA double null strain, suggesting that the encoded sensors contribute substantially to NarL V88A cross talk. Separately, the V88A substitution altered the in vitro rates of NarL autodephosphorylation and transmitter-stimulated dephosphorylation and decreased affinity for the cognate sensor, NarX. Together, these experiments show that the residue at position T + 1 can strongly influence two distinct aspects of receiver domain function, the autodephosphorylation rate and cross talk inhibition. IMPORTANCE: Many bacterial species contain a dozen or more discrete sensor-response regulator two-component systems that convert a specific input into a distinct output pattern. Cross talk, the unwanted transfer of signals between circuits, occurs when a response regulator is phosphorylated inappropriately from a noncognate source. Cross talk is inhibited in part by the high interaction specificity between cognate sensor-response regulator pairs. This study shows that a relatively subtle missense change from Val to Ala nullifies cross talk inhibition, enabling at least two noncognate sensors to enforce an inappropriate output independently of the relevant input.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/physiology , Mutation, Missense , Receptor Cross-Talk/physiology , Signal Transduction/genetics , Amino Acid Substitution , Carbamyl Phosphate/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Phosphorylation
20.
Sci Rep ; 5: 12892, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26256666

ABSTRACT

Pseudomonas syringae is a phytopathogenic bacterium widely spread on terrestrial plants. Sulfodiaminophosphinyl tripeptide Phaseolotoxins (PHTs), produced by P. syringae pv. phaseolicola and P. syringae pv. actinidiae, represent a kind of antimetabolic phytotoxins. PHTs inhibit host cell Ornithine transcarbamylase (OTCase) activity and induce Arginine auxotrophic phenotype. The biosynthesis of PHT is temperature dependent, being optically produced at around 18 °C, while blocked above 28 °C. PHT resistant OTCase ArgK acts as a functional replacement of housekeeping OTCase ArgF, which is the acting target of PHT, to confer PHT producers with self-resistance. It was postulated that argK might be regulated directly by a PHT biosynthetic precursor and indirectly by temperature with an unknown manner. Neither transcriptional regulator nor thermal regulation related protein encoding gene was detected from PHT biosynthetic gene cluster. The tripeptide, Cit-Ala-hArg, was identified to be a by-product of PHT biosynthetic pathway in this report. Formation of Cit-Ala-hArg was catalyzed by ArgK with tripeptide Orn-Ala-hArg and carbamyl phosphate as substrates. It showed that ArgK not only provided alternative Arginine source as reported previously, but also controlled the production of PHTs by converting PHT biosynthetic precursors to nontoxic Cit-Ala-hArg reservoir for producers' self-defense.


Subject(s)
Bacterial Proteins/metabolism , Ornithine Carbamoyltransferase/metabolism , Ornithine/analogs & derivatives , Pseudomonas syringae/enzymology , Amino Acid Sequence , Bacterial Proteins/classification , Bacterial Proteins/genetics , Carbamyl Phosphate/metabolism , Chromatography, High Pressure Liquid , Magnetic Resonance Spectroscopy , Mass Spectrometry , Multigene Family , Ornithine/chemistry , Ornithine/metabolism , Ornithine Carbamoyltransferase/classification , Ornithine Carbamoyltransferase/genetics , Phylogeny , Pseudomonas syringae/genetics , Pseudomonas syringae/metabolism , Substrate Specificity , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...