Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 534
Filtrer
1.
Plant Mol Biol ; 114(3): 60, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38758412

RÉSUMÉ

Pyruvate kinase (Pyk, EC 2.7.1.40) is a glycolytic enzyme that generates pyruvate and adenosine triphosphate (ATP) from phosphoenolpyruvate (PEP) and adenosine diphosphate (ADP), respectively. Pyk couples pyruvate and tricarboxylic acid metabolisms. Synechocystis sp. PCC 6803 possesses two pyk genes (encoded pyk1, sll0587 and pyk2, sll1275). A previous study suggested that pyk2 and not pyk1 is essential for cell viability; however, its biochemical analysis is yet to be performed. Herein, we biochemically analyzed Synechocystis Pyk2 (hereafter, SyPyk2). The optimum pH and temperature of SyPyk2 were 7.0 and 55 °C, respectively, and the Km values for PEP and ADP under optimal conditions were 1.5 and 0.053 mM, respectively. SyPyk2 is activated in the presence of glucose-6-phosphate (G6P) and ribose-5-phosphate (R5P); however, it remains unaltered in the presence of adenosine monophosphate (AMP) or fructose-1,6-bisphosphate. These results indicate that SyPyk2 is classified as PykA type rather than PykF, stimulated by sugar monophosphates, such as G6P and R5P, but not by AMP. SyPyk2, considering substrate affinity and effectors, can play pivotal roles in sugar catabolism under nonphotosynthetic conditions.


Sujet(s)
Glucose-6-phosphate , Phosphoénolpyruvate , Pyruvate kinase , Ribose monophosphate , Synechocystis , Synechocystis/métabolisme , Synechocystis/génétique , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Phosphoénolpyruvate/métabolisme , Glucose-6-phosphate/métabolisme , Ribose monophosphate/métabolisme , Spécificité du substrat , Concentration en ions d'hydrogène , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Cinétique , Température
2.
EMBO J ; 43(13): 2636-2660, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38778156

RÉSUMÉ

During infection viruses hijack host cell metabolism to promote their replication. Here, analysis of metabolite alterations in macrophages exposed to poly I:C recognises that the antiviral effector Protein Kinase RNA-activated (PKR) suppresses glucose breakdown within the pentose phosphate pathway (PPP). This pathway runs parallel to central glycolysis and is critical to producing NADPH and pentose precursors for nucleotides. Changes in metabolite levels between wild-type and PKR-ablated macrophages show that PKR controls the generation of ribose 5-phosphate, in a manner distinct from its established function in gene expression but dependent on its kinase activity. PKR phosphorylates and inhibits the Ribose 5-Phosphate Isomerase A (RPIA), thereby preventing interconversion of ribulose- to ribose 5-phosphate. This activity preserves redox control but decreases production of ribose 5-phosphate for nucleotide biosynthesis. Accordingly, the PKR-mediated immune response to RNA suppresses nucleic acid production. In line, pharmacological targeting of the PPP during infection decreases the replication of the Herpes simplex virus. These results identify an immune response-mediated control of host cell metabolism and suggest targeting the RPIA as a potential innovative antiviral treatment.


Sujet(s)
Macrophages , Voie des pentoses phosphates , Ribose monophosphate , eIF-2 Kinase , Animaux , Ribose monophosphate/métabolisme , Souris , eIF-2 Kinase/métabolisme , eIF-2 Kinase/génétique , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/virologie , Aldose-ketose isomerases/métabolisme , Aldose-ketose isomerases/génétique , ARN/métabolisme , ARN/génétique , Poly I-C/pharmacologie , Acides nucléiques/métabolisme , Acides nucléiques/immunologie , Réplication virale , Phosphorylation
3.
Nat Commun ; 15(1): 2999, 2024 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-38589375

RÉSUMÉ

Ribose-5-phosphate (R5P) is a precursor for nucleic acid biogenesis; however, the importance and homeostasis of R5P in the intracellular parasite Toxoplasma gondii remain enigmatic. Here, we show that the cytoplasmic sedoheptulose-1,7-bisphosphatase (SBPase) is dispensable. Still, its co-deletion with transaldolase (TAL) impairs the double mutant's growth and increases 13C-glucose-derived flux into pentose sugars via the transketolase (TKT) enzyme. Deletion of the latter protein affects the parasite's fitness but is not lethal and is correlated with an increased carbon flux via the oxidative pentose phosphate pathway. Further, loss of TKT leads to a decline in 13C incorporation into glycolysis and the TCA cycle, resulting in a decrease in ATP levels and the inability of phosphoribosyl-pyrophosphate synthetase (PRPS) to convert R5P into 5'-phosphoribosyl-pyrophosphate and thereby contribute to the production of AMP and IMP. Likewise, PRPS is essential for the lytic cycle. Not least, we show that RuPE-mediated metabolic compensation is imperative for the survival of the ΔsbpaseΔtal strain. In conclusion, we demonstrate that multiple routes can flexibly supply R5P to enable parasite growth and identify catalysis by TKT and PRPS as critical enzymatic steps. Our work provides novel biological and therapeutic insights into the network design principles of intracellular parasitism in a clinically-relevant pathogen.


Sujet(s)
Toxoplasma , Toxoplasma/métabolisme , Diphosphates/métabolisme , Ribose monophosphate/métabolisme , Glycolyse , Voie des pentoses phosphates
4.
Chembiochem ; 23(13): e202200147, 2022 07 05.
Article de Anglais | MEDLINE | ID: mdl-35476788

RÉSUMÉ

In nature 2-deoxy-D-ribose-5-phosphate aldolase (DERA) catalyses the reversible formation of 2-deoxyribose 5-phosphate from D-glyceraldehyde 3-phosphate and acetaldehyde. In addition, this enzyme can use acetaldehyde as the sole substrate, resulting in a tandem aldol reaction, yielding 2,4,6-trideoxy-D-erythro-hexapyranose, which spontaneously cyclizes. This reaction is very useful for the synthesis of the side chain of statin-type drugs used to decrease cholesterol levels in blood. One of the main challenges in the use of DERA in industrial processes, where high substrate loads are needed to achieve the desired productivity, is its inactivation by high acetaldehyde concentration. In this work, the utility of different variants of Pectobacterium atrosepticum DERA (PaDERA) as whole cell biocatalysts to synthesize 2-deoxyribose 5-phosphate and 2,4,6-trideoxy-D-erythro-hexapyranose was analysed. Under optimized conditions, E. coli BL21 (PaDERA C-His AA C49M) whole cells yields 99 % of both products. Furthermore, this enzyme is able to tolerate 500 mM acetaldehyde in a whole-cell experiment which makes it suitable for industrial applications.


Sujet(s)
Escherichia coli , Fructose bisphosphate aldolase , Acétaldéhyde , Aldehyde-lyases/composition chimique , Aldehyde-lyases/génétique , Pectobacterium , Ribose monophosphate
5.
Curr Protoc ; 2(1): e347, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-35050551

RÉSUMÉ

A simple and efficient method for the preparation of α-D-ribose 1-phosphate and 2-deoxy-α-D-ribose 1-phosphate, key intermediates in nucleoside metabolism and important starting compounds for the enzymatic synthesis of various modified nucleosides, has been proposed. It consists in near-irreversible enzymatic phosphorolysis of readily prepared hydroiodide salts of 7-methylguanosine and 7-methyl-2'-deoxyguanosine, respectively, in the presence of purine nucleoside phosphorylase. α-D-Ribose 1-phosphate and 2-deoxy-α-D-ribose 1-phosphate are obtained in near quantitative yields (by HPLC analysis) and 74%-94% yields after their isolation and purification. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Preparation of α-D-ribose 1-phosphate barium salt (4a) Alternate Protocol 1: Preparation of 2-deoxy-α-D-ribose 1-phosphate barium salt (4b) Basic Protocol 2: Preparation of α-D-ribose 1-phosphate bis(cyclohexylammonium) salt (5a) Alternate Protocol 2: Preparation of 2-deoxy-α-D-ribose 1-phosphate bis(cyclohexylammonium) salt (5b).


Sujet(s)
Désoxyguanosine , Désoxyguanosine/analogues et dérivés , Guanosine/analogues et dérivés , Ribose monophosphate
6.
PLoS Biol ; 19(12): e3001468, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34860829

RÉSUMÉ

The structure of the metabolic network is highly conserved, but we know little about its evolutionary origins. Key for explaining the early evolution of metabolism is solving a chicken-egg dilemma, which describes that enzymes are made from the very same molecules they produce. The recent discovery of several nonenzymatic reaction sequences that topologically resemble central metabolism has provided experimental support for a "metabolism first" theory, in which at least part of the extant metabolic network emerged on the basis of nonenzymatic reactions. But how could evolution kick-start on the basis of a metal catalyzed reaction sequence, and how could the structure of nonenzymatic reaction sequences be imprinted on the metabolic network to remain conserved for billions of years? We performed an in vitro screening where we add the simplest components of metabolic enzymes, proteinogenic amino acids, to a nonenzymatic, iron-driven reaction network that resembles glycolysis and the pentose phosphate pathway (PPP). We observe that the presence of the amino acids enhanced several of the nonenzymatic reactions. Particular attention was triggered by a reaction that resembles a rate-limiting step in the oxidative PPP. A prebiotically available, proteinogenic amino acid cysteine accelerated the formation of RNA nucleoside precursor ribose-5-phosphate from 6-phosphogluconate. We report that iron and cysteine interact and have additive effects on the reaction rate so that ribose-5-phosphate forms at high specificity under mild, metabolism typical temperature and environmental conditions. We speculate that accelerating effects of amino acids on rate-limiting nonenzymatic reactions could have facilitated a stepwise enzymatization of nonenzymatic reaction sequences, imprinting their structure on the evolving metabolic network.


Sujet(s)
Cystéine/métabolisme , Fer/métabolisme , Ribose monophosphate/métabolisme , Acides aminés/métabolisme , Catalyse , Cystéine/composition chimique , Évolution moléculaire , Glucose/métabolisme , Glycolyse/physiologie , Fer/composition chimique , Spectroscopie par résonance magnétique/méthodes , Voies et réseaux métaboliques/physiologie , Origine de la vie , Voie des pentoses phosphates/génétique , Voie des pentoses phosphates/physiologie
7.
Appl Microbiol Biotechnol ; 105(16-17): 6215-6228, 2021 Aug.
Article de Anglais | MEDLINE | ID: mdl-34410440

RÉSUMÉ

Deoxyribose-5-phosphate aldolases (DERAs, EC 4.1.2.4) are acetaldehyde-dependent, Class I aldolases catalyzing in nature a reversible aldol reaction between an acetaldehyde donor (C2 compound) and glyceraldehyde-3-phosphate acceptor (C3 compound, C3P) to generate deoxyribose-5-phosphate (C5 compound, DR5P). DERA enzymes have been found to accept also other types of aldehydes as their donor, and in particular as acceptor molecules. Consequently, DERA enzymes can be applied in C-C bond formation reactions to produce novel compounds, thus offering a versatile biocatalytic alternative for synthesis. DERA enzymes, found in all kingdoms of life, share a common TIM barrel fold despite the low overall sequence identity. The catalytic mechanism is well-studied and involves formation of a covalent enzyme-substrate intermediate. A number of protein engineering studies to optimize substrate specificity, enzyme efficiency, and stability of DERA aldolases have been published. These have employed various engineering strategies including structure-based design, directed evolution, and recently also machine learning-guided protein engineering. For application purposes, enzyme immobilization and usage of whole cell catalysis are preferred methods as they improve the overall performance of the biocatalytic processes, including often also the stability of the enzyme. Besides single-step enzymatic reactions, DERA aldolases have also been applied in multi-enzyme cascade reactions both in vitro and in vivo. The DERA-based applications range from synthesis of commodity chemicals and flavours to more complicated and high-value pharmaceutical compounds. KEY POINTS: • DERA aldolases are versatile biocatalysts able to make new C-C bonds. • Synthetic utility of DERAs has been improved by protein engineering approaches. • Computational methods are expected to speed up the future DERA engineering efforts.


Sujet(s)
Aldehyde-lyases , Ribose monophosphate , Aldehyde-lyases/génétique , Aldehyde-lyases/métabolisme , Fructose bisphosphate aldolase , Spécificité du substrat
8.
Med ; 2(6): 736-754, 2021 06 11.
Article de Anglais | MEDLINE | ID: mdl-34223403

RÉSUMÉ

BACKGROUND: Upregulated glucose metabolism is a common feature of tumors. Glucose can be broken down by either glycolysis or the oxidative pentose phosphate pathway (oxPPP). The relative usage within tumors of these catabolic pathways remains unclear. Similarly, the extent to which tumors make biomass precursors from glucose, versus take them up from the circulation, is incompletely defined. METHODS: We explore human triple negative breast cancer (TNBC) metabolism by isotope tracing with [1,2-13C]glucose, a tracer that differentiates glycolytic versus oxPPP catabolism and reveals glucose-driven anabolism. Patients enrolled in clinical trial NCT03457779 and received IV infusion of [1,2-13C]glucose during core biopsy of their primary TNBC. Tumor samples were analyzed for metabolite labeling by liquid chromatography-mass spectrometry (LC-MS). Genomic and proteomic analyses were performed and related to observed metabolic fluxes. FINDINGS: TNBC ferments glucose to lactate, with glycolysis dominant over the oxPPP. Most ribose phosphate is nevertheless produced by oxPPP. Glucose also feeds amino acid synthesis, including of serine, glycine, aspartate, glutamate, proline and glutamine (but not asparagine). Downstream in glycolysis, tumor pyruvate and lactate labeling exceeds that found in serum, indicating that lactate exchange via monocarboxylic transporters is less prevalent in human TNBC compared with most normal tissues or non-small cell lung cancer. CONCLUSIONS: Glucose directly feeds ribose phosphate, amino acid synthesis, lactate, and the TCA cycle locally within human breast tumors.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Tumeurs du sein triple-négatives , Acides aminés , Glucose/métabolisme , Humains , Acide lactique/métabolisme , Protéomique , Ribose monophosphate
9.
Biosci Rep ; 41(6)2021 06 25.
Article de Anglais | MEDLINE | ID: mdl-33973628

RÉSUMÉ

Carbohydrate metabolism in heart failure shares similarities to that following hypoxic exposure, and is thought to maintain energy homoeostasis in the face of reduced O2 availability. As part of these in vivo adaptations during sustained hypoxia, the heart up-regulates and maintains a high glycolytic flux, but the underlying mechanism is still elusive. We followed the cardiac glycolytic responses to a chronic hypoxic (CH) intervention using [5-3H]-glucose labelling in combination with detailed and extensive enzymatic and metabolomic approaches to provide evidence of the underlying mechanism that allows heart survivability. Following 3 weeks of in vivo hypoxia (11% oxygen), murine hearts were isolated and perfused in a retrograde mode with function measured via an intraventricular balloon and glycolytic flux quantified using [5-3H]-glucose labelling. At the end of perfusion, hearts were flash-frozen and central carbon intermediates determined via liquid chromatography tandem mass spectrometry (LC-MS/MS). The maximal activity of glycolytic enzymes considered rate-limiting was assessed enzymatically, and protein abundance was determined using Western blotting. Relative to normoxic hearts, CH increased ex vivo cardiac glycolytic flux 1.7-fold with no effect on cardiac function. CH up-regulated cardiac pyruvate kinase (PK) flux 3.1-fold and cardiac pyruvate kinase muscle isoenzyme M2 (PKM2) protein content 1.4-fold compared with normoxic hearts. CH also augmented cardiac pentose phosphate pathway (PPP) flux, reflected by higher ribose-5-phosphate (R5P) content. These findings support an increase in the covalent (protein expression) and allosteric (flux) control of PKM2 as being central to the sustained up-regulation of the glycolytic flux in the chronically hypoxic heart.


Sujet(s)
Glycolyse , Hypoxie/enzymologie , Myocytes cardiaques/enzymologie , Pyruvate kinase/métabolisme , Régulation allostérique , Animaux , Maladie chronique , Modèles animaux de maladie humaine , Hypoxie/anatomopathologie , Préparation de coeur isolé , Mâle , Métabolome , Souris , Myocytes cardiaques/anatomopathologie , Voie des pentoses phosphates , Ribose monophosphate/métabolisme , Transduction du signal
10.
Antimicrob Agents Chemother ; 65(7): e0189220, 2021 06 17.
Article de Anglais | MEDLINE | ID: mdl-33875438

RÉSUMÉ

Neglected tropical diseases caused by kinetoplastid parasites (Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp.) place a significant health and economic burden on developing nations worldwide. Current therapies are largely outdated, inadequate, and face mounting drug resistance from the causative parasites. Thus, there is an urgent need for drug discovery and development. Target-led drug discovery approaches have focused on the identification of parasite enzymes catalyzing essential biochemical processes, which significantly differ from equivalent proteins found in humans, thereby providing potentially exploitable therapeutic windows. One such target is ribose 5-phosphate isomerase B (RpiB), an enzyme involved in the nonoxidative branch of the pentose phosphate pathway, which catalyzes the interconversion of d-ribose 5-phosphate and d-ribulose 5-phosphate. Although protozoan RpiB has been the focus of numerous targeted studies, compounds capable of selectively inhibiting this parasite enzyme have not been identified. Here, we present the results of a fragment library screening against Leishmania infantum RpiB (LiRpiB), performed using thermal shift analysis. Hit fragments were shown to be effective inhibitors of LiRpiB in activity assays, and several fragments were capable of selectively inhibiting parasite growth in vitro. These results support the identification of LiRpiB as a validated therapeutic target. The X-ray crystal structure of apo LiRpiB was also solved, permitting docking studies to assess how hit fragments might interact with LiRpiB to inhibit its activity. Overall, this work will guide structure-based development of LiRpiB inhibitors as antileishmanial agents.


Sujet(s)
Leishmania infantum , Préparations pharmaceutiques , Séquence d'acides aminés , Humains , Ribose monophosphate
11.
Nucleic Acids Res ; 49(1): 257-268, 2021 01 11.
Article de Anglais | MEDLINE | ID: mdl-33290564

RÉSUMÉ

8-Oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodGuo) is a biomarker of oxidative DNA damage and can be repaired by hOGG1 and APE1 via the base excision repair (BER) pathway. In this work, we studied coordinated BER of 8-oxodGuo by hOGG1 and APE1 in nucleosome core particles and found that histones transiently formed DNA-protein cross-links (DPCs) with active repair intermediates such as 3'-phospho-α,ß-unsaturated aldehyde (PUA) and 5'-deoxyribosephosphate (dRP). The effects of histone participation could be beneficial or deleterious to the BER process, depending on the circumstances. In the absence of APE1, histones enhanced the AP lyase activity of hOGG1 by cross-linking with 3'-PUA. However, the formed histone-PUA DPCs hampered the subsequent repair process. In the presence of APE1, both the AP lyase activity of hOGG1 and the formation of histone-PUA DPCs were suppressed. In this case, histones could catalyse removal of the 5'-dRP by transiently cross-linking with the active intermediate. That is, histones promoted the repair by acting as 5'-dRP lyases. Our findings demonstrate that histones participate in multiple steps of 8-oxodGuo repair in nucleosome core particles, highlighting the diverse roles that histones may play during DNA repair in eukaryotic cells.


Sujet(s)
8-Hydroxy-2'-désoxyguanosine/métabolisme , Réparation de l'ADN/physiologie , Histone/physiologie , Nucléosomes/métabolisme , Phosphorus-oxygen lyases/métabolisme , DNA Glycosylases/métabolisme , DNA-(apurinic or apyrimidinic site) lyase/métabolisme , Humains , Modèles moléculaires , Conformation d'acide nucléique , Nucléosomes/ultrastructure , Conformation des protéines , Ribose monophosphate/métabolisme
12.
RNA ; 26(12): 1838-1846, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-32843366

RÉSUMÉ

The Fibro-purF motif is a putative structured noncoding RNA domain that was discovered previously in species of Fibrobacter by using comparative sequence analysis methods. An updated bioinformatics search yielded a total of only 30 unique-sequence representatives, exclusively found upstream of the purF gene that codes for the enzyme amidophosphoribosyltransferase. This enzyme synthesizes the compound 5-phospho-D-ribosylamine (PRA), which is the first committed step in purine biosynthesis. The consensus model for Fibro-purF motif RNAs includes a predicted three-stem junction that carries numerous conserved nucleotide positions within the regions joining the stems. This architecture appears to be of sufficient size and complexity for the formation of the ligand-binding aptamer portion of a riboswitch. In this study, we conducted biochemical analyses of a representative Fibro-purF motif RNA to confirm that the RNA generally folds according to the predicted consensus model. However, due to the instability of PRA, binding of this ligand candidate by the RNA could not be directly assessed. Genetic analyses were used to demonstrate that Fibro-purF motif RNAs regulate gene expression in accordance with predicted PRA concentrations. These findings indicate that Fibro-purF motif RNAs are genetic regulation elements that likely suppress PRA biosynthesis when sufficient levels of this purine precursor are present.


Sujet(s)
Bactéries/métabolisme , Protéines bactériennes/métabolisme , Régulation de l'expression des gènes bactériens , Motifs nucléotidiques/génétique , ARN bactérien/métabolisme , Ribose monophosphate/métabolisme , Bactéries/génétique , Bactéries/croissance et développement , Protéines bactériennes/génétique , Séquence nucléotidique , ARN bactérien/composition chimique , ARN bactérien/génétique , Riborégulateur , Similitude de séquences
13.
Cell ; 182(4): 933-946.e14, 2020 08 20.
Article de Anglais | MEDLINE | ID: mdl-32780992

RÉSUMÉ

Methanol, being electron rich and derivable from methane or CO2, is a potentially renewable one-carbon (C1) feedstock for microorganisms. Although the ribulose monophosphate (RuMP) cycle used by methylotrophs to assimilate methanol differs from the typical sugar metabolism by only three enzymes, turning a non-methylotrophic organism to a synthetic methylotroph that grows to a high cell density has been challenging. Here we reprogrammed E. coli using metabolic robustness criteria followed by laboratory evolution to establish a strain that can efficiently utilize methanol as the sole carbon source. This synthetic methylotroph alleviated a so far uncharacterized hurdle, DNA-protein crosslinking (DPC), by insertion sequence (IS)-mediated copy number variations (CNVs) and balanced the metabolic flux by mutations. Being capable of growing at a rate comparable with natural methylotrophs in a wide range of methanol concentrations, this synthetic methylotrophic strain illustrates genome editing and evolution for microbial tropism changes and expands the scope of biological C1 conversion.


Sujet(s)
Escherichia coli/métabolisme , Génie métabolique , Méthanol/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Carbone/métabolisme , Cycle citrique/génétique , Variations de nombre de copies de segment d'ADN , Évolution moléculaire dirigée , Escherichia coli/génétique , Formaldéhyde/métabolisme , Glycolyse , Mutagenèse , Ribose monophosphate/métabolisme
14.
Appl Microbiol Biotechnol ; 104(15): 6429-6441, 2020 Aug.
Article de Anglais | MEDLINE | ID: mdl-32533303

RÉSUMÉ

Ribose-5-phosphate isomerase (Rpi, EC 5.3.1.6) is widespread in microorganisms, animals, and plants. It has a pivotal role in the pentose phosphate pathway and responsible for catalyzing the isomerization between D-ribulose 5-phosphate and D-ribose 5-phosphate. In recent years, Rpi has received considerable attention as a multipurpose biocatalyst for production of rare sugars, including D-allose, L-rhamnulose, L-lyxose, and L-tagatose. Besides, it has been thought of as a potential drug target in the treatment of trypanosomatid-caused diseases such as Chagas' disease, leishmaniasis, and human African trypanosomiasis. Despite increased research activities, up to now, no systematic review of Rpi has been published. To fill this gap, this paper provides detailed information about the enzymatic properties of various Rpis. Furthermore, structural features, catalytic mechanism, and molecular modifications of Rpis are summarized based on extensive crystal structure research. Additionally, the applications of Rpi in rare sugar production and the role of Rpi in trypanocidal drug design are reviewed. Key points • Fundamental properties of various ribose-5-phosphate isomerases (Rpis). • Differences in crystal structure and catalytic mechanism between RpiA and RpiB. • Application of Rpi as a rare sugar producer and a potential drug target.


Sujet(s)
Aldose-ketose isomerases/composition chimique , Aldose-ketose isomerases/métabolisme , Aldose-ketose isomerases/classification , Animaux , Sites de fixation , Biocatalyse , Cristallographie aux rayons X , Humains , Isomérie , Cinétique , Modèles moléculaires , Maladies parasitaires/traitement médicamenteux , Plantes/enzymologie , Ribose monophosphate/métabolisme
15.
Sci Rep ; 10(1): 9051, 2020 06 03.
Article de Anglais | MEDLINE | ID: mdl-32493959

RÉSUMÉ

Uridine phosphorylase (UP) is a key enzyme of pyrimidine salvage pathways that enables the recycling of endogenous or exogenous-supplied pyrimidines and plays an important intracellular metabolic role. Here, we biochemically and structurally characterized two evolutionarily divergent uridine phosphorylases, PcUP1 and PcUP2 from the oomycete pathogen Phytophthora capsici. Our analysis of other oomycete genomes revealed that both uridine phosphorylases are present in Phytophthora and Pythium genomes, but only UP2 is seen in Saprolegnia spp. which are basal members of the oomycetes. Moreover, uridine phosphorylases are not found in obligate oomycete pathogens such as Hyaloperonospora arabidopsidis and Albugo spp. PcUP1 and PcUP2 are upregulated 300 and 500 fold respectively, within 90 min after infection of pepper leaves. The crystal structures of PcUP1 in ligand-free and in complex with uracil/ribose-1-phosphate, 2'-deoxyuridine/phosphate and thymidine/phosphate were analyzed. Crystal structure of this uridine phosphorylase showed strict conservation of key residues in the binding pocket. Structure analysis of PcUP1 with bound ligands, and site-directed mutagenesis of key residues provide additional support for the "push-pull" model of catalysis. Our study highlights the importance of pyrimidine salvage during the earliest stages of infection.


Sujet(s)
Phytophthora/métabolisme , Uridine phosphorylase/composition chimique , Uridine phosphorylase/métabolisme , Sites de fixation/physiologie , Catalyse , Domaine catalytique/physiologie , Cristallographie aux rayons X/méthodes , Désoxyuridine/composition chimique , Désoxyuridine/métabolisme , Ligands , Pyrimidines/composition chimique , Pyrimidines/métabolisme , Ribose monophosphate/composition chimique , Ribose monophosphate/métabolisme , Thymidine/composition chimique , Thymidine/métabolisme , Uracile/composition chimique , Uracile/métabolisme , Uridine/composition chimique , Uridine/métabolisme
16.
RNA ; 26(5): 613-628, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32111664

RÉSUMÉ

Functions of eukaryotic mRNAs are characterized by intramolecular interactions between their ends. We have addressed the question whether 5' and 3' ends meet by diffusion-controlled encounter "through solution" or by a mechanism involving the RNA backbone. For this purpose, we used a translation system derived from Drosophila embryos that displays two types of 5'-3' interactions: Cap-dependent translation initiation is stimulated by the poly(A) tail and inhibited by Smaug recognition elements (SREs) in the 3' UTR. Chimeric RNAs were made consisting of one RNA molecule carrying a luciferase coding sequence and a second molecule containing SREs and a poly(A) tail; the two were connected via a protein linker. The poly(A) tail stimulated translation of such chimeras even when disruption of the RNA backbone was combined with an inversion of the 5'-3' polarity between the open reading frame and poly(A) segment. Stimulation by the poly(A) tail also decreased with increasing RNA length. Both observations suggest that contacts between the poly(A) tail and the 5' end are established through solution, independently of the RNA backbone. In the same chimeric constructs, SRE-dependent inhibition of translation was also insensitive to disruption of the RNA backbone. Thus, tracking of the backbone is not involved in the repression of cap-dependent initiation. However, SRE-dependent repression was insensitive to mRNA length, suggesting that the contact between the SREs in the 3' UTR and the 5' end of the RNA might be established in a manner that differs from the contact between the poly(A) tail and the cap.


Sujet(s)
Stabilité de l'ARN/génétique , ARN messager/génétique , ARN/génétique , Ribose/composition chimique , Régions 3' non traduites/génétique , Régions 5' non traduites/génétique , Cellules eucaryotes , Cadres ouverts de lecture/génétique , Poly A/génétique , Biosynthèse des protéines/génétique , Coiffes des ARN/génétique , Ribose/génétique , Ribose monophosphate/composition chimique , Ribose monophosphate/génétique
17.
Oxid Med Cell Longev ; 2020: 5367102, 2020.
Article de Anglais | MEDLINE | ID: mdl-32104534

RÉSUMÉ

Treatments on neoplastic diseases and cancer using genotoxic drugs often cause long-term health problems related to premature aging. The underlying mechanism is poorly understood. Based on the study of a long-lasting senescence-like growth arrest (10-12 weeks) of human dermal fibroblasts induced by psoralen plus UVA (PUVA) treatment, we here revealed that slowly repaired bulky DNA damages can serve as a "molecular scar" leading to reduced cell proliferation through persistent endogenous production of reactive oxygen species (ROS) that caused accelerated telomere erosion. The elevated levels of ROS were the results of mitochondrial dysfunction and the activation of NADPH oxidase (NOX). A combined inhibition of DNA-PK and PARP1 could suppress the level of ROS. Together with a reduced expression level of BRCA1 as well as the upregulation of PP2A and 53BP1, these data suggest that the NHEJ repair of DNA double-strand breaks may be the initial trigger of metabolic changes leading to ROS production. Further study showed that stimulation of the pentose phosphate pathway played an important role for NOX activation, and ROS could be efficiently suppressed by modulating the NADP/NADPH ratio. Interestingly, feeding cells with ribose-5-phosphate, a precursor for nucleotide biosynthesis that produced through the PPP, could evidently suppress the ROS level and prevent the cell enlargement related to mitochondrial biogenesis. Taken together, these results revealed an important signaling pathway between DNA damage repair and the cell metabolism, which contributed to the premature aging effects of PUVA, and may be generally applicable for a large category of chemotherapeutic reagents including many cancer drugs.


Sujet(s)
Vieillissement de la cellule/physiologie , Altération de l'ADN/physiologie , Stress oxydatif/physiologie , Cellules cultivées , Vieillissement de la cellule/génétique , Altération de l'ADN/génétique , Réparation de l'ADN/génétique , Réparation de l'ADN/physiologie , Humains , NADP/génétique , NADP/métabolisme , Oxydoréduction , Stress oxydatif/génétique , Poly (ADP-Ribose) polymerase-1/génétique , Poly (ADP-Ribose) polymerase-1/métabolisme , Espèces réactives de l'oxygène/métabolisme , Ribose monophosphate/métabolisme , Protéine-1 liant le suppresseur de tumeur p53/génétique , Protéine-1 liant le suppresseur de tumeur p53/métabolisme
18.
J Med Chem ; 63(6): 3238-3251, 2020 03 26.
Article de Anglais | MEDLINE | ID: mdl-32052631

RÉSUMÉ

Analogues of the Ca2+-releasing intracellular messenger d-myo-inositol 1,4,5-trisphosphate [1, Ins(1,4,5)P3] are important synthetic targets. Replacement of the α-glucopyranosyl motif in the natural product mimic adenophostin 2 by d-chiro-inositol in d-chiro-inositol adenophostin 4 increased the potency. Similar modification of the non-nucleotide Ins(1,4,5)P3 mimic ribophostin 6 may increase the activity. d-chiro-Inositol ribophostin 10 was synthesized by coupling as building blocks suitably protected ribose 12 with l-(+)-3-O-trifluoromethylsulfonyl-6-O-p-methoxybenzyl-1,2:4,5-di-O-isopropylidene-myo-inositol 11. Separable diastereoisomeric 3-O-camphanate esters of (±)-6-O-p-methoxy-benzyl-1,2:4,5-di-O-isopropylidene-myo-inositol allowed the preparation of 11. Selective trans-isopropylidene deprotection in coupled 13, then monobenzylation gave separable regioisomers 15 and 16. p-Methoxybenzyl group deprotection of 16, phosphitylation/oxidation, then deprotection afforded 10, which was a full agonist in Ca2+-release assays; its potency and binding affinity for Ins(1,4,5)P3R were similar to those of adenophostin. Both 4 and 10 elicited a store-operated Ca2+ current ICRAC in patch-clamped cells, unlike Ins(1,4,5)P3 consistent with resistance to metabolism. d-chiro-Inositol ribophostin is the most potent small-molecule Ins(1,4,5)P3 receptor agonist without a nucleobase yet synthesized.


Sujet(s)
Récepteurs à l'inositol 1,4,5-triphosphate/agonistes , Inositol phosphates/pharmacologie , Ribose monophosphate/pharmacologie , Animaux , Calcium/métabolisme , Lignée cellulaire , Poulets , Humains , Inositol phosphates/synthèse chimique , Structure moléculaire , Rats , Ribose monophosphate/synthèse chimique , Relation structure-activité
19.
Sci Rep ; 10(1): 3397, 2020 02 25.
Article de Anglais | MEDLINE | ID: mdl-32099005

RÉSUMÉ

Collagen fibrils are central to the molecular organization of the extracellular matrix (ECM) and to defining the cellular microenvironment. Glycation of collagen fibrils is known to impact on cell adhesion and migration in the context of cancer and in model studies, glycation of collagen molecules has been shown to affect the binding of other ECM components to collagen. Here we use TEM to show that ribose-5-phosphate (R5P) glycation of collagen fibrils - potentially important in the microenvironment of actively dividing cells, such as cancer cells - disrupts the longitudinal ordering of the molecules in collagen fibrils and, using KFM and FLiM, that R5P-glycated collagen fibrils have a more negative surface charge than unglycated fibrils. Altered molecular arrangement can be expected to impact on the accessibility of cell adhesion sites and altered fibril surface charge on the integrity of the extracellular matrix structure surrounding glycated collagen fibrils. Both effects are highly relevant for cell adhesion and migration within the tumour microenvironment.


Sujet(s)
Collagène de type I/composition chimique , Matrice extracellulaire/composition chimique , Ribose monophosphate/composition chimique , Animaux , Collagène de type I/métabolisme , Matrice extracellulaire/métabolisme , Glycosylation , Humains , Ribose monophosphate/métabolisme
20.
J Biol Chem ; 294(35): 13061-13072, 2019 08 30.
Article de Anglais | MEDLINE | ID: mdl-31300556

RÉSUMÉ

The presence of ribonucleoside monophosphates (rNMPs) in nuclear DNA decreases genome stability. To ensure survival despite rNMP insertions, cells have evolved a complex network of DNA repair mechanisms, in which the ribonucleotide excision repair pathway, initiated by type 2 RNase H (RNase HII/2), plays a major role. We recently demonstrated that eukaryotic RNase H2 cannot repair damage, that is, ribose monophosphate abasic (both apurinic or apyrimidinic) site (rAP) or oxidized rNMP embedded in DNA. Currently, it remains unclear why RNase H2 is unable to repair these modified nucleic acids having either only a sugar moiety or an oxidized base. Here, we compared the endoribonuclease specificity of the RNase HII enzymes from the archaeon Pyrococcus abyssi and the bacterium Escherichia coli, examining their ability to process damaged rNMPs embedded in DNA in vitro We found that E. coli RNase HII cleaves both rAP and oxidized rNMP sites. In contrast, like the eukaryotic RNase H2, P. abyssi RNase HII did not display any rAP or oxidized rNMP incision activities, even though it recognized them. Notably, the archaeal enzyme was also inactive on a mismatched rNMP, whereas the E. coli enzyme displayed a strong preference for the mispaired rNMP over the paired rNMP in DNA. On the basis of our biochemical findings and also structural modeling analyses of RNase HII/2 proteins from organisms belonging to all three domains of life, we propose that RNases HII/2's dual roles in ribonucleotide excision repair and RNA/DNA hydrolysis result in limited acceptance of modified rNMPs embedded in DNA.


Sujet(s)
ADN/métabolisme , Escherichia coli/métabolisme , Ribonuclease H/métabolisme , Ribonucléotides/métabolisme , Ribose monophosphate/métabolisme , Cellules HeLa , Humains , Oxydoréduction , Cellules cancéreuses en culture
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE