Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 404
Filter
1.
PLoS Pathog ; 17(4): e1009350, 2021 04.
Article in English | MEDLINE | ID: mdl-33878133

ABSTRACT

Intestinal epithelial cells are subject to attack by a diverse array of microbes, including intracellular as well as extracellular pathogens. While defense in epithelial cells can be triggered by pattern recognition receptor-mediated detection of microbe-associated molecular patterns, there is much to be learned about how they sense infection via perturbations of host physiology, which often occur during infection. A recently described host defense response in the nematode C. elegans called the Intracellular Pathogen Response (IPR) can be triggered by infection with diverse natural intracellular pathogens, as well as by perturbations to protein homeostasis. From a forward genetic screen, we identified the C. elegans ortholog of purine nucleoside phosphorylase pnp-1 as a negative regulator of IPR gene expression, as well as a negative regulator of genes induced by extracellular pathogens. Accordingly, pnp-1 mutants have resistance to both intracellular and extracellular pathogens. Metabolomics analysis indicates that C. elegans pnp-1 likely has enzymatic activity similar to its human ortholog, serving to convert purine nucleosides into free bases. Classic genetic studies have shown how mutations in human purine nucleoside phosphorylase cause immunodeficiency due to T-cell dysfunction. Here we show that C. elegans pnp-1 acts in intestinal epithelial cells to regulate defense. Altogether, these results indicate that perturbations in purine metabolism are likely monitored as a cue to promote defense against epithelial infection in the nematode C. elegans.


Subject(s)
Epithelial Cells/metabolism , Purine Nucleosides/metabolism , Purine-Nucleoside Phosphorylase/genetics , Receptors, Pattern Recognition/metabolism , Animals , Bacterial Infections/prevention & control , Caenorhabditis elegans/metabolism , Cell Count/methods , Purine-Nucleoside Phosphorylase/deficiency
2.
Int J Mol Sci ; 23(23)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36499470

ABSTRACT

Purine nucleosides represent an interesting group of nitrogen heterocycles, showing a wide range of biological effects. In this study, we designed and synthesized a series of 6,9-disubstituted and 2,6,9-trisubstituted purine ribonucleosides via consecutive nucleophilic aromatic substitution, glycosylation, and deprotection of the ribofuranose unit. We prepared eight new purine nucleosides bearing unique adamantylated aromatic amines at position 6. Additionally, the ability of the synthesized purine nucleosides to form stable host-guest complexes with ß-cyclodextrin (ß-CD) was confirmed using nuclear magnetic resonance (NMR) and mass spectrometry (ESI-MS) experiments. The in vitro antiproliferative activity of purine nucleosides and their equimolar mixtures with ß-CD was tested against two types of human tumor cell line. Six adamantane-based purine nucleosides showed an antiproliferative activity in the micromolar range. Moreover, their effect was only slightly suppressed by the presence of ß-CD, which was probably due to the competitive binding of the corresponding purine nucleoside inside the ß-CD cavity.


Subject(s)
Adamantane , beta-Cyclodextrins , Humans , Adamantane/pharmacology , Purine Nucleosides/pharmacology , Purine Nucleosides/metabolism , beta-Cyclodextrins/pharmacology , Cell Line, Tumor , Nucleosides/pharmacology , Nucleosides/chemistry
3.
Biopolymers ; 112(1): e23399, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32969496

ABSTRACT

The notion of using synthetic heterocycles instead of the native bases to interface with DNA and RNA has been explored for nearly 60 years. Unnatural bases compatible with the DNA/RNA coding interface have the potential to expand the genetic code and co-opt the machinery of biology to access new macromolecular function; accordingly, this body of research is core to synthetic biology. While much of the literature on artificial bases focuses on code expansion, there is a significant and growing effort on docking synthetic heterocycles to noncoding nucleic acid interfaces; this approach seeks to illuminate major processes of nucleic acids, including regulation of transcription, translation, transport, and transcript lifetimes. These major avenues of research at the coding and noncoding interfaces have in common fundamental principles in molecular recognition. Herein, we provide an overview of foundational literature in biophysics of base recognition and unnatural bases in coding to provide context for the developing area of targeting noncoding nucleic acid interfaces with synthetic bases, with a focus on systems developed through iterative design and biophysical study.


Subject(s)
DNA/metabolism , RNA/metabolism , Base Pairing , DNA/chemistry , Hydrogen Bonding , Purine Nucleosides/chemistry , Purine Nucleosides/metabolism , Pyrimidine Nucleosides/chemistry , Pyrimidine Nucleosides/metabolism , RNA/chemistry , Synthetic Biology/methods
4.
Nucleic Acids Res ; 46(7): 3458-3467, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29438559

ABSTRACT

Reactive oxygen species (ROS) are formed in mitochondria during electron transport and energy generation. Elevated levels of ROS lead to increased amounts of mitochondrial DNA (mtDNA) damage. We report that levels of M1dG, a major endogenous peroxidation-derived DNA adduct, are 50-100-fold higher in mtDNA than in nuclear DNA in several different human cell lines. Treatment of cells with agents that either increase or decrease mitochondrial superoxide levels leads to increased or decreased levels of M1dG in mtDNA, respectively. Sequence analysis of adducted mtDNA suggests that M1dG residues are randomly distributed throughout the mitochondrial genome. Basal levels of M1dG in mtDNA from pulmonary microvascular endothelial cells (PMVECs) from transgenic bone morphogenetic protein receptor 2 mutant mice (BMPR2R899X) (four adducts per 106 dG) are twice as high as adduct levels in wild-type cells. A similar increase was observed in mtDNA from heterozygous null (BMPR2+/-) compared to wild-type PMVECs. Pulmonary arterial hypertension is observed in the presence of BMPR2 signaling disruptions, which are also associated with mitochondrial dysfunction and oxidant injury to endothelial tissue. Persistence of M1dG adducts in mtDNA could have implications for mutagenesis and mitochondrial gene expression, thereby contributing to the role of mitochondrial dysfunction in diseases.


Subject(s)
DNA, Mitochondrial/metabolism , Mitochondria/genetics , Oxidative Stress/genetics , Purine Nucleosides/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , DNA Adducts/genetics , DNA Adducts/metabolism , DNA, Mitochondrial/genetics , Electron Transport/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/genetics , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Lipid Peroxidation/genetics , Mice , Mice, Transgenic , Mitochondria/pathology , Mutagenesis/genetics , Oxidants/pharmacology , Purine Nucleosides/biosynthesis , Reactive Oxygen Species/chemistry , Superoxides/metabolism
5.
Molecules ; 25(5)2020 Mar 09.
Article in English | MEDLINE | ID: mdl-32182773

ABSTRACT

The bi-enzymatic synthesis of the antiviral drug vidarabine (arabinosyladenine, ara-A), catalyzed by uridine phosphorylase from Clostridium perfringens (CpUP) and a purine nucleoside phosphorylase from Aeromonas hydrophila (AhPNP), was re-designed under continuous-flow conditions. Glyoxyl-agarose and EziGTM1 (Opal) were used as immobilization carriers for carrying out this preparative biotransformation. Upon setting-up reaction parameters (substrate concentration and molar ratio, temperature, pressure, residence time), 1 g of vidarabine was obtained in 55% isolated yield and >99% purity by simply running the flow reactor for 1 week and then collecting (by filtration) the nucleoside precipitated out of the exiting flow. Taking into account the substrate specificity of CpUP and AhPNP, the results obtained pave the way to the use of the CpUP/AhPNP-based bioreactor for the preparation of other purine nucleosides.


Subject(s)
Antiviral Agents/chemistry , Enzymes, Immobilized/chemistry , Purine-Nucleoside Phosphorylase/chemistry , Vidarabine/chemistry , Aeromonas hydrophila/enzymology , Biocatalysis , Bioreactors , Biotransformation/drug effects , Clostridium perfringens/enzymology , Enzymes, Immobilized/genetics , Glyoxylates/chemistry , Humans , Protein Engineering/methods , Purine Nucleosides/chemistry , Purine Nucleosides/metabolism , Purine-Nucleoside Phosphorylase/genetics , Sepharose/chemistry , Substrate Specificity , Vidarabine/biosynthesis , Vidarabine/genetics
6.
J Biol Inorg Chem ; 24(7): 985-997, 2019 10.
Article in English | MEDLINE | ID: mdl-31359185

ABSTRACT

Nucleobases serve as ideal targets where drugs bind and exert their anticancer activities. Cisplatin (cisPt) preferentially coordinates to 2'-deoxyguanosine (dGuo) residues within DNA. The dGuo adducts that are formed alter the DNA structure, contributing to inhibition of function and ultimately cancer cell death. Despite its success as an anticancer drug, cisPt has a number of drawbacks that reduce its efficacy, including repair of adducts and drug resistance. Some approaches to overcome this problem involve development of compounds that coordinate to other purine nucleobases, including those found in RNA. In this work, amino acid-linked platinum(II) (AAPt) compounds of alanine and ornithine (AlaPt and OrnPt, respectively) were studied. Their reactivity preferences for DNA and RNA purine nucleosides (i.e., 2'-deoxyadenosine (dAdo), adenosine (Ado), dGuo, and guanosine (Guo)) were determined. The chosen compounds form predominantly monofunctional adducts by reacting at the N1, N3, or N7 positions of purine nucleobases. In addition, features of AAPt compounds that impact the glycosidic bond stability of Ado residues were explored. The glycosidic bond cleavage is activated differentially for AlaPt-Ado and OrnPt-Ado isomers. Formation of unique adducts at non-canonical residues and subsequent destabilization of the glycosidic bonds are important features that could circumvent platinum-based drug resistance.


Subject(s)
Alanine/chemistry , Glycosides/chemistry , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/metabolism , Ornithine/chemistry , Ornithine/metabolism , Purine Nucleosides/metabolism , Purine Nucleosides/chemistry
7.
Semin Immunol ; 27(3): 177-83, 2015 May.
Article in English | MEDLINE | ID: mdl-25818531

ABSTRACT

Inflammatory diseases in mucosal organs as diverse as the lung, liver and intestine inevitably require the intimate interactions between neutrophils and epithelia. The physiologic consequences of such interactions often determine endpoint organ function, and for this reason, much recent interest has developed in identifying mechanisms and novel targets to promote the resolution of mucosal inflammation. Physiologically-relevant in vitro and in vivo model systems have aided in discovery of novel pathways to define basic inflammatory mechanisms and approaches to defining the concepts of inflammatory resolution. Here, we will review the recent literature regarding the contribution of neutrophils to inflammatory resolution, with an emphasis on the role of the tissue microenvironment, endogenous pathways for promoting resolution and the molecular determinants of neutrophil-epithelial cell interactions during ongoing inflammation. These recent studies highlight the dynamic nature of pro-resolving pathways and lend insight into the complexity of treating mucosal inflammation.


Subject(s)
Epithelial Cells/immunology , Homeostasis/immunology , Inflammation/immunology , Mucous Membrane/immunology , Neutrophils/immunology , Cell Communication/immunology , Cell Hypoxia/immunology , Cell Movement/immunology , Cellular Microenvironment/immunology , Humans , Mucous Membrane/cytology , Mucous Membrane/pathology , Oxygen Consumption/immunology , Oxygen Consumption/physiology , Purine Nucleosides/metabolism
8.
Int J Mol Sci ; 20(9)2019 May 07.
Article in English | MEDLINE | ID: mdl-31067671

ABSTRACT

Effects of fructose 1,6-bisphosphate (F-1,6-P2) towards N-methyl-d-aspartate NMDA excitotoxicity were evaluated in rat organotypic hippocampal brain slice cultures (OHSC) challenged for 3 h with 30 µM NMDA, followed by incubations (24, 48, and 72 h) without (controls) and with F-1,6-P2 (0.5, 1 or 1.5 mM). At each time, cell necrosis was determined by measuring LDH in the medium. Energy metabolism was evaluated by measuring ATP, GTP, ADP, AMP, and ATP catabolites (nucleosides and oxypurines) in deproteinized OHSC extracts. Gene expressions of phosphofructokinase, aldolase, and glyceraldehyde-3-phosphate dehydrogenase were also measured. F-1,6-P2 dose-dependently decreased NMDA excitotoxicity, abolishing cell necrosis at the highest concentration tested (1.5 mM). Additionally, F-1,6-P2 attenuated cell energy imbalance caused by NMDA, ameliorating the mitochondrial phosphorylating capacity (increase in ATP/ADP ratio) Metabolism normalization occurred when using 1.5 mM F-1,6-P2. Remarkable increase in expressions of phosphofructokinase, aldolase and glyceraldehyde-3-phosphate dehydrogenase (up to 25 times over the values of controls) was also observed. Since this phenomenon was recorded even in OHSC treated with F-1,6-P2 with no prior challenge with NMDA, it is highly conceivable that F-1,6-P2 can enter into intact cerebral cells producing significant benefits on energy metabolism. These effects are possibly mediated by changes occurring at the gene level, thus opening new perspectives for F-1,6-P2 application as a useful adjuvant to rescue mitochondrial metabolism of cerebral cells under stressing conditions.


Subject(s)
Fructose-Bisphosphatase/pharmacology , Hippocampus/drug effects , N-Methylaspartate/toxicity , Neuroprotective Agents/pharmacology , Animals , Energy Metabolism , Fructose-Bisphosphate Aldolase/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Necrosis , Phosphofructokinases/metabolism , Purine Nucleosides/metabolism , Rats , Rats, Wistar
9.
Molecules ; 25(1)2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31888088

ABSTRACT

Purine nucleoside phosphorylases (PNPs) are promising biocatalysts for the synthesis of purine nucleoside analogs. Although a number of PNPs have been reported, the development of highly efficient enzymes for industrial applications is still in high demand. Herein, a new trimeric purine nucleoside phosphorylase (AmPNP) from Aneurinibacillus migulanus AM007 was cloned and heterologously expressed in Escherichia coli BL21(DE3). The AmPNP showed good thermostability and a broad range of pH stability. The enzyme was thermostable below 55 °C for 12 h (retaining nearly 100% of its initial activity), and retained nearly 100% of the initial activity in alkaline buffer systems (pH 7.0-9.0) at 60 °C for 2 h. Then, a one-pot, two-enzyme mode of transglycosylation reaction was successfully constructed by combining pyrimidine nucleoside phosphorylase (BbPyNP) derived from Brevibacillus borstelensis LK01 and AmPNP for the production of purine nucleoside analogs. Conversions of 2,6-diaminopurine ribonucleoside (1), 2-amino-6-chloropurine ribonucleoside (2), and 6-thioguanine ribonucleoside (3) synthesized still reached >90% on the higher concentrations of substrates (pentofuranosyl donor: purine base; 20:10 mM) with a low enzyme ratio of BbPyNP: AmPNP (2:20 µg/mL). Thus, the new trimeric AmPNP is a promising biocatalyst for industrial production of purine nucleoside analogs.


Subject(s)
Bacillales/enzymology , Purine Nucleosides/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Bacillales/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Cloning, Molecular , Enzyme Stability , Purine-Nucleoside Phosphorylase/chemistry , Purine-Nucleoside Phosphorylase/genetics , Pyrimidine Phosphorylases/metabolism , Thermodynamics
10.
Org Biomol Chem ; 16(32): 5800-5807, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30063056

ABSTRACT

We synthesized a small library of eighteen 5-substituted pyrimidine or 7-substituted 7-deazapurine nucleoside triphosphates bearing methyl, ethynyl, phenyl, benzofuryl or dibenzofuryl groups through cross-coupling reactions of nucleosides followed by triphosphorylation or through direct cross-coupling reactions of halogenated nucleoside triphosphates. We systematically studied the influence of the modification on the efficiency of T7 RNA polymerase catalyzed synthesis of modified RNA and found that modified ATP, UTP and CTP analogues bearing smaller modifications were good substrates and building blocks for the RNA synthesis even in difficult sequences incorporating multiple modified nucleotides. Bulky dibenzofuryl derivatives of ATP and GTP were not substrates for the RNA polymerase. In the case of modified GTP analogues, a modified procedure using a special promoter and GMP as initiator needed to be used to obtain efficient RNA synthesis. The T7 RNA polymerase synthesis of modified RNA can be very efficiently used for synthesis of modified RNA but the method has constraints in the sequence of the first three nucleotides of the transcript, which must contain a non-modified G in the +1 position.


Subject(s)
Bacteriophage T7/enzymology , DNA-Directed RNA Polymerases/metabolism , Purine Nucleosides/metabolism , Purines/metabolism , Pyrimidine Nucleosides/metabolism , RNA/metabolism , Viral Proteins/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Cytidine Triphosphate/analogs & derivatives , Cytidine Triphosphate/metabolism , Purine Nucleosides/chemistry , Purines/chemistry , Pyrimidine Nucleosides/chemistry , RNA/chemistry , Substrate Specificity , Uridine Triphosphate/analogs & derivatives , Uridine Triphosphate/metabolism
11.
Br Poult Sci ; 58(5): 536-543, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28547997

ABSTRACT

1. This study was conducted to evaluate the effects of purine nucleosides on performance, gut morphology, intestinal enzymes and immunity functions in broiler chickens from 0 to 21 d of age. 2. A total of 360 1-d-old male chickens (Cobb 500) were randomly assigned to 4 treatments with 6 replications. Experimental diets consisted of a control without any additives and diets containing 0.1% pure adenosine, 0.1% pure guanosine and 0.1% equal aliquots of pure adenosine and guanosine. Two birds per cage (12 birds per treatment) were killed on d 11 and 21 in order to obtain serum samples for lipid profile, jejunal samples for morphology and mucosal immunity, digestive enzymes for epithelial maturation, and bursa and spleen samples for relative weight of immune organs to live body weight. 3. Birds receiving adenosine in their diets showed a significant increase in body weight and average daily gain and a significantly lower feed conversion ratio compared to the control birds. Villus height and width in jejunal samples also increased significantly in birds supplemented with adenosine. Although maltase was not affected by the experimental diets, adenosine increased alkaline phosphatase and aminopeptidase. Adenosine and its combination with guanosine boosted mucosal immunity as a result of increased IgA production. While there was no significant difference among treatments regarding the relative weight of the spleen, adenosine increased the relative weight of the bursa of Fabricius. Present results also showed that adding guanosine to broiler diets had no significant effects on growth, gut morphology, enzymes activity and immunological indices. 4. In conclusion, the improvement in growth performance, gut morphology and immunity in birds receiving adenosine demonstrated that pure adenosine could be a beneficial feed additive for the poultry industry, while guanosine showed no significant improvement.


Subject(s)
Chickens/physiology , Immunity, Mucosal/drug effects , Intestines/physiology , Purine Nucleosides/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Bursa of Fabricius/physiology , Chickens/anatomy & histology , Chickens/growth & development , Chickens/immunology , Diet/veterinary , Dietary Supplements/analysis , Intestinal Mucosa/immunology , Intestines/enzymology , Intestines/growth & development , Male , Organ Size , Purine Nucleosides/administration & dosage , Random Allocation , Spleen/physiology
12.
Br J Nutr ; 113(4): 560-73, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25619278

ABSTRACT

About 20 % of ruminal microbial N in dairy cows derives from purines and pyrimidines; however, their intermediary metabolism and contribution to the overall N metabolism has sparsely been described. In the present study, the postprandial patterns of net portal-drained viscera (PDV) and hepatic metabolism were assessed to evaluate purine and pyrimidine N in dairy cows. Blood was sampled simultaneously from four veins with eight hourly samples from four multi-catheterised Holstein cows. Quantification of twenty purines and pyrimidines was performed with HPLC-MS/MS, and net fluxes were estimated across the PDV, hepatic tissue and total splanchnic tissue (TSP). Concentration differences between veins of fifteen purine and pyrimidine nucleosides (NS), bases (BS) and degradation products (DP) were different from zero (P≤ 0·05), resulting in the net PDV releases of purine NS (0·33-1·3 mmol/h), purine BS (0·0023-0·018 mmol/h), purine DP (7·0-7·8 mmol/h), pyrimidine NS (0·30-2·8 mmol/h) and pyrimidine DP (0·047-0·77 mmol/h). The hepatic removal of purine and pyrimidine was almost equivalent to the net PDV release, resulting in no net TSP release. One exception was uric acid (7·9 mmol/h) from which a large net TSP release originated from the degradation of purine NS and BS. A small net TSP release of the pyrimidine DP ß-alanine and ß-aminoisobutyric acid (-0·032 to 0·37 mmol/h) demonstrated an outlet of N into the circulating N pool. No effect of time relative to feeding was observed (P>0·05). These data indicate that considerable amounts of N are lost in the dairy cow due to prominent intermediary degradation of purines, but that pyrimidine N is reusable to a larger extent.


Subject(s)
Intestinal Absorption , Lactation/metabolism , Liver/metabolism , Nitrogen Cycle , Purines/metabolism , Pyrimidines/metabolism , Secondary Metabolism , Animals , Animals, Inbred Strains , Cattle , Dairying , Digestion , Female , Hydrolysis , Lactation/blood , Postprandial Period , Purine Nucleosides/blood , Purine Nucleosides/metabolism , Purines/blood , Pyrimidine Nucleosides/blood , Pyrimidine Nucleosides/metabolism , Pyrimidines/blood , Random Allocation , Spleen/metabolism , Uric Acid/blood , Uric Acid/metabolism
13.
Eukaryot Cell ; 13(5): 572-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24585883

ABSTRACT

The intracellular pathogen Toxoplasma gondii is a purine auxotroph that relies on purine salvage for proliferation. We have optimized T. gondii purine nucleoside phosphorylase (TgPNP) stability and crystallized TgPNP with phosphate and immucillin-H, a transition-state analogue that has high affinity for the enzyme. Immucillin-H bound to TgPNP with a dissociation constant of 370 pM, the highest affinity of 11 immucillins selected to probe the catalytic site. The specificity for transition-state analogues indicated an early dissociative transition state for TgPNP. Compared to Plasmodium falciparum PNP, large substituents surrounding the 5'-hydroxyl group of inhibitors demonstrate reduced capacity for TgPNP inhibition. Catalytic discrimination against large 5' groups is consistent with the inability of TgPNP to catalyze the phosphorolysis of 5'-methylthioinosine to hypoxanthine. In contrast to mammalian PNP, the 2'-hydroxyl group is crucial for inhibitor binding in the catalytic site of TgPNP. This first crystal structure of TgPNP describes the basis for discrimination against 5'-methylthioinosine and similarly 5'-hydroxy-substituted immucillins; structural differences reflect the unique adaptations of purine salvage pathways of Apicomplexa.


Subject(s)
Enzyme Inhibitors/chemistry , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Purine-Nucleoside Phosphorylase/chemistry , Purine-Nucleoside Phosphorylase/metabolism , Toxoplasma/enzymology , Catalysis , Catalytic Domain , Crystallography, X-Ray , Kinetics , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Purine Nucleosides/chemistry , Purine Nucleosides/metabolism , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/genetics , Pyrimidinones/chemistry , Substrate Specificity , Toxoplasma/chemistry , Toxoplasma/genetics
14.
Chem Pharm Bull (Tokyo) ; 63(2): 134-42, 2015.
Article in English | MEDLINE | ID: mdl-25748785

ABSTRACT

A number of new 2,6-disubstituted-1-deazanebularine analogues as well as two structurally related pyrazole-fused tricyclic nucleosides were prepared. Their synthesis was carried out by the conversion of 6-amino-2-picoline to a suitable 1-deazapurine, followed by a Vorbrüggen type glycosylation and subsequent elaboration of the condensed pyrazole ring. The synthesized nebularine analogues proved to be weak adenosine deaminase inhibitors.


Subject(s)
Adenosine Deaminase Inhibitors/chemical synthesis , Adenosine Deaminase/chemistry , Purine Nucleosides/chemistry , Ribonucleosides/chemistry , Adenosine Deaminase/metabolism , Adenosine Deaminase Inhibitors/chemistry , Adenosine Deaminase Inhibitors/metabolism , Animals , Cattle , Glycosylation , Magnetic Resonance Spectroscopy , Protein Binding , Purine Nucleosides/chemical synthesis , Purine Nucleosides/metabolism , Pyrazoles/chemistry , Ribonucleosides/chemical synthesis , Ribonucleosides/metabolism
15.
Alcohol Clin Exp Res ; 38(6): 1550-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24891074

ABSTRACT

BACKGROUND: Alcohol use disorders are often associated with lung disease. Alcohol exposure leads to the production of reactive oxygen species, lipid peroxidation, and formation of malondialdehyde (MDA) as well as to induce the expression of cytochrome p450 2E1 (CYP2E1). Likewise, cigarette smoking can lead to lung lipid peroxidation and formation of MDA. MDA can bind to DNA forming MDA-deoxyguanosine (M1dG) adducts, which have been implicated in alcohol-related cancers and cardiovascular disease. Because CYP2E1 regulates MDA production, and our previous studies have shown that alcohol and cigarette smoke can lead to MDA formation, we hypothesized that CYP2E1 would modulate M1dG adduct formation and single-strand DNA damage in alcohol- and cigarette smoke-exposed lung cells and tissue. METHODS: Normal human bronchial epithelial cells (HBECs) were pretreated with 10 µM diallyl disulfide (DADS) for 1 hour and treated with 80 mM ethanol (EtOH) ± 5% cigarette smoke extract (CSE) for 3 hours for comet assay and 6 hours for CYP2E1, MDA, and M1dG adduct assays. C57BL/6 mice were administered 20% EtOH ad libitum in drinking water for 8 weeks and exposed to whole-body cigarette smoke for 5 weeks. Mice were also fed a CYP2E1 inhibitor, DADS, at 1 µM/g of feed in their daily diet for 7 weeks. Whole lung tissue homogenate was used for CYP2E1, MDA, and M1dG adduct assays. RESULTS: EtOH exposure significantly increased HBEC olive tail moment. DADS pretreatment of HBECs attenuated this EtOH effect. EtOH also induced MDA and M1dG adduct formation, which was also significantly reduced by DADS treatment. CSE ± EtOH did not enhance these effects. In lung tissue homogenate of 8-week alcohol-fed mice, MDA and M1dG adduct levels were significantly elevated in comparison with control mice and mice fed DADS while consuming alcohol. No increase in MDA and M1dG adduct formation was observed in 5-week cigarette smoke-exposed mice. CONCLUSIONS: These findings suggest that CYP2E1 plays a pivotal role in alcohol-induced M1dG adducts, and the use of DADS as dietary supplement can reverse the effects of alcohol on M1dG formation.


Subject(s)
Allyl Compounds/pharmacology , Cytochrome P-450 CYP2E1 Inhibitors/pharmacology , Disulfides/pharmacology , Ethanol/pharmacology , Purine Nucleosides/metabolism , Animals , Cells, Cultured , DNA Damage/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice, Inbred C57BL , Respiratory Mucosa , Smoking/adverse effects , Smoking/metabolism
16.
Semin Cancer Biol ; 22(4): 335-41, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22414910

ABSTRACT

About a century ago Otto Warburg observed that tumor cells exhibited increased glycolysis despite the presence of oxygen and stated this metabolic shift to glycolysis as the origin of cancer cell. In the meantime it has become clear, that the altered glucose metabolism is only one piece of the tumor metabolome puzzle. In addition, amino acid, lipid and adenosine metabolism are adapted to fulfill the tumors needs for energy and generation of building blocks such as lipids and nucleotides for new cell structures. The altered tumor metabolism leads to accumulation of specific metabolites in the tumor environment and creates a favorable milieu for tumor growth, progression and metastasis. These tumor-derived metabolites are important players in immune escape mechanisms beside other known factors such as cytokines, chemokines and growth factors. A variety of metabolites re-educate immune cells and prevent an effective immune response against tumor cells. Furthermore, tumor infiltrating immune cells support tumor growth by the secretion of cytokines, growth factors and other metabolic determinants. Hence, a complex interplay of tumor metabolites, cytokines and stromal factors is active in tumors and facilitates their establishment and growth. Pharmacological blockade of tumor metabolites could overcome some limitations of cancer treatment and rescue the endogenous immune response against tumor cells.


Subject(s)
Immunomodulation , Neoplasms/metabolism , Acidosis , Amino Acids/metabolism , Animals , Biological Transport , Disease Progression , Glycolysis , Humans , Lactic Acid/metabolism , Mitochondria/metabolism , Neoplasms/immunology , Neoplasms/pathology , Pentose Phosphate Pathway , Prostaglandin-Endoperoxide Synthases/metabolism , Purine Nucleosides/metabolism
17.
PLoS One ; 19(2): e0293378, 2024.
Article in English | MEDLINE | ID: mdl-38386624

ABSTRACT

This study evaluated 15 lactic acid bacteria with a focus on their ability to degrade inosine and hypo-xanthine-which are the intermediates in purine metabolism-for the management of hyperuricemia and gout. After a preliminary screening based on HPLC, Lactiplantibacillus plantarum CR1 and Lactiplantibacillus pentosus GZ1 were found to have the highest nucleoside degrading rates, and they were therefore selected for further characterization. S. thermophilus IDCC 2201, which possessed the hpt gene encoding hypoxanthine-guanine phosphoribosyltransferase (HGPRT) and exhibited purine degradation, was also selected for further characterization. These three selected strains were examined in terms of their probiotic effect on lowering serum uric acid in a Sprague-Dawley (SD) rat model of potassium oxonate (PO)-induced hyperuricemia. Among these three strains, the level of serum uric acid was most reduced by S. thermophilus IDCC 2201 (p < 0.05). Further, analysis of the microbiome showed that administration of S. thermophlilus IDCC 2201 led to a significant difference in gut microbiota composition compared to that in the group administered with PO-induced hyperuricemia. Moreover, intestinal short-chain fatty acids (SCFAs) were found to be significantly increased. Altogether, the results of this work indicate that S. thermophilus IDCC 2201 lowers uric acid levels by degrading purine-nucleosides and also restores intestinal flora and SCFAs, ultimately suggesting that S. thermophilus IDCC 2201 is a promising candidate for use as an adjuvant treatment in patients with hyperuricemia.


Subject(s)
Hyperuricemia , Purine Nucleosides , Rats , Animals , Humans , Purine Nucleosides/metabolism , Uric Acid , Hyperuricemia/metabolism , Nucleosides , Streptococcus thermophilus , Rats, Sprague-Dawley , Xanthine
18.
Biochemistry ; 52(37): 6525-36, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23972005

ABSTRACT

Proteins of unknown function belonging to cog1816 and cog0402 were characterized. Sav2595 from Steptomyces avermitilis MA-4680, Acel0264 from Acidothermus cellulolyticus 11B, Nis0429 from Nitratiruptor sp. SB155-2 and Dr0824 from Deinococcus radiodurans R1 were cloned, purified, and their substrate profiles determined. These enzymes were previously incorrectly annotated as adenosine deaminases or chlorohydrolases. It was shown here that these enzymes actually deaminate 6-aminodeoxyfutalosine. The deamination of 6-aminodeoxyfutalosine is part of an alternative menaquinone biosynthetic pathway that involves the formation of futalosine. 6-Aminodeoxyfutalosine is deaminated by these enzymes with catalytic efficiencies greater than 10(5) M(-1) s(-1), Km values of 0.9-6.0 µM, and kcat values of 1.2-8.6 s(-1). Adenosine, 2'-deoxyadenosine, thiomethyladenosine, and S-adenosylhomocysteine are deaminated at least an order of magnitude slower than 6-aminodeoxyfutalosine. The crystal structure of Nis0429 was determined and the substrate, 6-aminodeoxyfutalosine, was positioned in the active site on the basis of the presence of adventitiously bound benzoic acid. In this model, Ser-145 interacts with the carboxylate moiety of the substrate. The structure of Dr0824 was also determined, but a collapsed active site pocket prevented docking of substrates. A computational model of Sav2595 was built on the basis of the crystal structure of adenosine deaminase and substrates were docked. The model predicted a conserved arginine after ß-strand 1 to be partially responsible for the substrate specificity of Sav2595.


Subject(s)
Nucleoside Deaminases/metabolism , Purine Nucleosides/metabolism , Vitamin K 2/metabolism , Actinomycetales/enzymology , Catalytic Domain , Crystallography, X-Ray , Deamination , Deinococcus/enzymology , Epsilonproteobacteria/enzymology , Epsilonproteobacteria/genetics , Kinetics , Models, Molecular , Molecular Docking Simulation , Nucleoside Deaminases/genetics , Streptomyces/enzymology , Streptomyces/genetics , Substrate Specificity
19.
Mol Pharmacol ; 84(6): 865-75, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24068428

ABSTRACT

A mycoplasma-encoded purine nucleoside phosphorylase (designated PNPHyor) has been cloned and characterized for the first time. Efficient phosphorolysis of natural 6-oxopurine and 6-aminopurine nucleosides was observed, with adenosine the preferred natural substrate (Km = 61 µM). Several cytostatic purine nucleoside analogs proved to be susceptible to PNPHyor-mediated phosphorolysis, and a markedly decreased or increased cytostatic activity was observed in Mycoplasma hyorhinis-infected human breast carcinoma MCF-7 cell cultures (MCF-7.Hyor), depending on the properties of the released purine base. We demonstrated an ∼10-fold loss of cytostatic activity of cladribine in MCF-7.Hyor cells and observed a rapid and complete phosphorolysis of this drug when it was exposed to the supernatant of mycoplasma-infected cells. This conversion (inactivation) could be prevented by a specific PNP inhibitor. These findings correlated well with the high efficiency of PNPHyor-catalyzed phosphorolysis of cladribine to its less toxic base 2-chloroadenine (Km = 80 µM). In contrast, the cytostatic activity of nucleoside analogs carrying a highly toxic purine base and being a substrate for PNPHyor, but not human PNP, was substantially increased in MCF-7.Hyor cells (∼130-fold for fludarabine and ∼45-fold for 6-methylpurine-2'-deoxyriboside). Elimination of the mycoplasma from the tumor cell cultures or selective inhibition of PNPHyor by a PNP inhibitor restored the cytostatic activity of the purine-based nucleoside drugs. Since several studies suggest a high and preferential colonization or association of tumor tissue in cancer patients with different prokaryotes (including mycoplasmas), the data presented here may be of relevance for the optimization of purine nucleoside-based anticancer drug treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Mycoplasma hyorhinis/enzymology , Purine-Nucleoside Phosphorylase/metabolism , Purines/pharmacology , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cladribine/metabolism , Cladribine/pharmacology , Drug Screening Assays, Antitumor , Humans , Kinetics , Mycoplasma hyorhinis/genetics , Purine Nucleosides/metabolism , Purine Nucleosides/pharmacology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/genetics , Purines/metabolism , Structure-Activity Relationship , Substrate Specificity
20.
J Pharmacol Exp Ther ; 345(3): 331-41, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23504005

ABSTRACT

The presence and activity of nucleotides and dinucleotides in the physiology of most, if not all, organisms, from bacteria to humans, have been recognized by the scientific community, and the eye is no exception. Nucleotides in the dynamic fluids interact with many ocular structures, such as the tears and aqueous humor. Moreover, high concentrations of nucleotides in these secretions may reflect disease states such as dry eye and glaucoma. Apart from the nucleotide concentration in these fluids, P2 purinergic receptors have been described on the ocular surface (cornea and conjunctiva), anterior pole (ciliary body, trabecular meshwork), and posterior pole (retina). P2X and P2Y purinergic receptors are essential in maintaining the homeostasis of ocular processes, such as tear secretion, aqueous humor production, or retinal modulation. When they are functioning properly, they allow the eye to do its job (to see), but in some cases, a lack or an excess of nucleotides or a malfunction in the corresponding purinergic receptors leads to disease. This Perspective is focused on the nucleotides and dinucleotides and the P2 purinergic receptors in the eye and how they contribute to normal and disease states. We also emphasize the action of nucleotides and their receptors and antagonists as potential therapeutic agents.


Subject(s)
Eye Diseases/drug therapy , Nucleotides/physiology , Ocular Physiological Phenomena , Animals , Aqueous Humor/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Eye/immunology , Eye Diseases/metabolism , Humans , Nucleotides/metabolism , Purine Nucleosides/metabolism , Purine Nucleosides/physiology , Purine Nucleotides/metabolism , Purine Nucleotides/physiology , Retina/drug effects , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/physiology , Tears/chemistry , Tears/metabolism , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL