scholarly journals The Rut Pathway for Pyrimidine Degradation: Novel Chemistry and Toxicity Problems

2010 ◽  
Vol 193 (1) ◽  
pp. 326-326
Author(s):  
K.-S. Kim ◽  
J. G. Pelton ◽  
W. B. Inwood ◽  
U. Andersen ◽  
S. Kustu ◽  
...  
1970 ◽  
Vol 245 (21) ◽  
pp. 5668-5676
Author(s):  
Yoshikazu Sanno ◽  
Mary Holzer ◽  
Robert T. Schimke

2019 ◽  
Vol 48 (4) ◽  
pp. 1886-1904 ◽  
Author(s):  
Jihane Basbous ◽  
Antoine Aze ◽  
Laurent Chaloin ◽  
Rana Lebdy ◽  
Dana Hodroj ◽  
...  

Abstract Imbalance in the level of the pyrimidine degradation products dihydrouracil and dihydrothymine is associated with cellular transformation and cancer progression. Dihydropyrimidines are degraded by dihydropyrimidinase (DHP), a zinc metalloenzyme that is upregulated in solid tumors but not in the corresponding normal tissues. How dihydropyrimidine metabolites affect cellular phenotypes remains elusive. Here we show that the accumulation of dihydropyrimidines induces the formation of DNA–protein crosslinks (DPCs) and causes DNA replication and transcriptional stress. We used Xenopus egg extracts to recapitulate DNA replication invitro. We found that dihydropyrimidines interfere directly with the replication of both plasmid and chromosomal DNA. Furthermore, we show that the plant flavonoid dihydromyricetin inhibits human DHP activity. Cellular exposure to dihydromyricetin triggered DPCs-dependent DNA replication stress in cancer cells. This study defines dihydropyrimidines as potentially cytotoxic metabolites that may offer an opportunity for therapeutic-targeting of DHP activity in solid tumors.


2004 ◽  
Vol 379 (1) ◽  
pp. 119-124 ◽  
Author(s):  
André B. P. van KUILENBURG ◽  
Alida E. M. STROOMER ◽  
Henk van LENTHE ◽  
Nico G. G. M. ABELING ◽  
Albert H. van GENNIP

DPD (dihydropyrimidine dehydrogenase) constitutes the first step of the pyrimidine degradation pathway, in which the pyrimidine bases uracil and thymine are catabolized to β-alanine and the R-enantiomer of β-AIB (β-aminoisobutyric acid) respectively. The S-enantiomer of β-AIB is predominantly derived from the catabolism of valine. It has been suggested that an altered homoeostasis of β-alanine underlies some of the clinical abnormalities encountered in patients with a DPD deficiency. In the present study, we demonstrated that only a slightly decreased concentration of β-alanine was present in the urine and plasma, whereas normal levels of β-alanine were present in the cerebrospinal fluid of patients with a DPD deficiency. Therefore the metabolism of β-alanine-containing peptides, such as carnosine, may be an important factor involved in the homoeostasis of β-alanine in patients with DPD deficiency. The mean concentration of β-AIB was approx. 2–3-fold lower in cerebrospinal fluid and urine of patients with a DPD deficiency, when compared with controls. In contrast, strongly decreased levels (10-fold) of β-AIB were present in the plasma of DPD patients. Our results demonstrate that, under pathological conditions, the catabolism of valine can result in the production of significant amounts of β-AIB. Furthermore, the observation that the R-enantiomer of β-AIB is abundantly present in the urine of DPD patients suggests that significant cross-over exists between the thymine and valine catabolic pathways.


Metabolism ◽  
1997 ◽  
Vol 46 (11) ◽  
pp. 1339-1342 ◽  
Author(s):  
Tetsuya Yamamoto ◽  
Yuji Moriwaki ◽  
Sumio Takahashi ◽  
Zenta Tsutsumi ◽  
Jun-ichi Yamakita ◽  
...  

2010 ◽  
Vol 192 (16) ◽  
pp. 4089-4102 ◽  
Author(s):  
Kwang-Seo Kim ◽  
Jeffrey G. Pelton ◽  
William B. Inwood ◽  
Ulla Andersen ◽  
Sydney Kustu ◽  
...  

ABSTRACT The Rut pathway is composed of seven proteins, all of which are required by Escherichia coli K-12 to grow on uracil as the sole nitrogen source. The RutA and RutB proteins are central: no spontaneous suppressors arise in strains lacking them. RutA works in conjunction with a flavin reductase (RutF or a substitute) to catalyze a novel reaction. It directly cleaves the uracil ring between N-3 and C-4 to yield ureidoacrylate, as established by both nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry. Although ureidoacrylate appears to arise by hydrolysis, the requirements for the reaction and the incorporation of 18O at C-4 from molecular oxygen indicate otherwise. Mass spectrometry revealed the presence of a small amount of product with the mass of ureidoacrylate peracid in reaction mixtures, and we infer that this is the direct product of RutA. In vitro RutB cleaves ureidoacrylate hydrolytically to release 2 mol of ammonium, malonic semialdehyde, and carbon dioxide. Presumably the direct products are aminoacrylate and carbamate, both of which hydrolyze spontaneously. Together with bioinformatic predictions and published crystal structures, genetic and physiological studies allow us to predict functions for RutC, -D, and -E. In vivo we postulate that RutB hydrolyzes the peracid of ureidoacrylate to yield the peracid of aminoacrylate. We speculate that RutC reduces aminoacrylate peracid to aminoacrylate and RutD increases the rate of spontaneous hydrolysis of aminoacrylate. The function of RutE appears to be the same as that of YdfG, which reduces malonic semialdehyde to 3-hydroxypropionic acid. RutG appears to be a uracil transporter.


2019 ◽  
Author(s):  
Mariana López-Sámano ◽  
Luis Fernando Lozano-Aguirre Beltrán ◽  
Rosina Sánchez-Thomas ◽  
Araceli Dávalos ◽  
Tomás Villaseñor ◽  
...  

Abstractβ-alanine synthesis in bacteria occurs by the decarboxylation of L-aspartate as part of the pantothenate synthesis pathway. In the other two domains of life we find different pathways for β-alanine formation, such as sources from spermine in plants, uracil in yeast and by transamination reactions in insects and mammals. There are also promiscuous decarboxylases that can decarboxylate aspartate. Several bioinformatics studies about the conservation of pantothenate synthesis pathway performed on bacteria, archaea and eukaryotes, have shown a partial conservation of the pathway. As a part of our work, we performed an analysis of the prevalence of reported β-alanine synthesis pathways in 204 genomes of alpha-proteobacteria, with a focus on theRhizobialesorder. The aim of this work was to determine the enzyme or pathway used to synthetize β-alanine inRhizobium etliCFN42. Our bioinformatics analysis showed that this strain encodes the pyrimidine degradation pathway in its genome. We obtained a β-alanine synthase (amaB)mutant that was a β-alanine auxotroph. Complementation with the cloned gene restored the wild type phenotype. Biochemical analysis confirmed that the recombinant AmaB catalyzed the formation of β-alanine from 3-Ureidopropionic acidin vitro. Here we show a different way in bacteria to produce this essential metabolite.ImportanceSince the pioneer studies of Cronan (1980) on β-alanine synthesis inE. coli, it has been assumed that the decarboxilation of aspartate by the L-aspartate-α-decarboxylase it’s the main enzymatic reaction for β-alanine synthesis in bacteria. Forty years later, while we were studying the pantothenic acid synthesis in rhizobia, we demonstrate that a numerous and diverse group of bacteria classified as α-proteobacteria synthesize β-alaninede novousing β-alanine synthase, the last enzyme from the reductive pathway for uracil degradation.Additionally, there is a growing interest in β-amino acid due to its remarkable pharmaceuticals properties as hypoglycemic, antiketogenic and anti-fungal agents.


2000 ◽  
Vol 46 (12) ◽  
pp. 1916-1922 ◽  
Author(s):  
Henk van Lenthe ◽  
André B P van Kuilenburg ◽  
Tetsuya Ito ◽  
Albert H Bootsma ◽  
Arno van Cruchten ◽  
...  

Abstract Background: Urinary concentrations of thymine, uracil, and their degradation products are useful indicators of deficiencies of enzymes of the pyrimidine degradation pathway. We describe a rapid, specific method to measure these concentrations to detect inborn errors of pyrimidine metabolism. Methods: We used urine or urine-soaked filter-paper strips as samples and measured thymine, uracil, and their degradation products dihydrothymine, dihydrouracil, N-carbamyl-β-aminoisobutyric acid, and N-carbamyl-β-alanine. Reversed-phase HPLC was combined with electrospray ionization tandem mass spectrometry, and detection was performed by multiple-reaction monitoring. Stable-isotope-labeled reference compounds were used as internal standards. Results: All pyrimidine degradation products could be measured in one analytical run of 15 min. Detection limits were 0.4–4 μmol/L. The intraassay imprecision (CV) of urine samples with added compounds was 1.3–12% for liquid urines and 1.0–10% for filter-paper extracts of the urines. The interassay imprecision (CV) was 3–11% (100–200 μmol/L). Recoveries were 89–99% at 100–200 μmol/L and 95–106% at 1 mmol/L in liquid urines, and 93–103% at 100–200 μmol/L and 100–106% at 1 mmol/L in filter-paper samples. Correct identifications of deficiencies of the pyrimidine-degrading enzymes were readily made with urine samples from patients with known defects. Conclusions: HPLC with electrospray ionization tandem mass spectrometry allows rapid testing for disorders of the pyrimidine degradation pathway, and filter-paper samples allow easy collection, transport, and storage of urine samples.


1997 ◽  
Vol 20 (2) ◽  
pp. 203-213 ◽  
Author(s):  
A.H. van Gennip ◽  
N.G.G.M. Abeling ◽  
P. Vreken ◽  
A.B.P. van Kuilenburg

Sign in / Sign up

Export Citation Format

Share Document