Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Nucleic Acids Res ; 48(7): 3975-3986, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32095808

ABSTRACT

Guanine-rich regions of the human genome can adopt non-canonical secondary structures. Their role in regulating gene expression has turned them into promising targets for therapeutic intervention. Ligands based on polyaromatic moieties are especially suitable for targeting G-quadruplexes utilizing their size complementarity to interact with the large exposed surface area of four guanine bases. A predictable way of (de)stabilizing specific G-quadruplex structures through efficient base stacking of polyaromatic functional groups could become a valuable tool in our therapeutic arsenal. We have investigated the effect of pyrene-modified uridine nucleotides incorporated at several positions of the thrombin binding aptamer (TBA) as a model system. Characterization using spectroscopic and biophysical methods provided important insights into modes of interaction between pyrene groups and the G-quadruplex core as well as (de)stabilization by enthalpic and entropic contributions. NMR data demonstrated that incorporation of pyrene group into G-rich oligonucleotide such as TBA may result in significant changes in 3D structure such as formation of novel dimeric topology. Site specific structural changes induced by stacking of the pyrene moiety on nearby nucleobases corelate with distinct thrombin binding affinities and increased resistance against nuclease degradation.


Subject(s)
Aptamers, Nucleotide/chemistry , G-Quadruplexes , Pyrenes/chemistry , Aptamers, Nucleotide/blood , Aptamers, Nucleotide/metabolism , Deoxyribonucleases , Dimerization , Entropy , Humans , Thermodynamics , Thrombin/metabolism , Uracil Nucleotides/chemistry
2.
Nature ; 514(7521): 252-256, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25119025

ABSTRACT

The pluripotency factor Lin28 inhibits the biogenesis of the let-7 family of mammalian microRNAs. Lin28 is highly expressed in embryonic stem cells and has a fundamental role in regulation of development, glucose metabolism and tissue regeneration. Overexpression of Lin28 is correlated with the onset of numerous cancers, whereas let-7, a tumour suppressor, silences several human oncogenes. Lin28 binds to precursor let-7 (pre-let-7) hairpins, triggering the 3' oligo-uridylation activity of TUT4 and TUT7 (refs 10-12). The oligoU tail added to pre-let-7 serves as a decay signal, as it is rapidly degraded by Dis3l2 (refs 13, 14), a homologue of the catalytic subunit of the RNA exosome. The molecular basis of Lin28-mediated recruitment of TUT4 and TUT7 to pre-let-7 and its subsequent degradation by Dis3l2 is largely unknown. To examine the mechanism of Dis3l2 substrate recognition we determined the structure of mouse Dis3l2 in complex with an oligoU RNA to mimic the uridylated tail of pre-let-7. Three RNA-binding domains form an open funnel on one face of the catalytic domain that allows RNA to navigate a path to the active site different from that of its exosome counterpart. The resulting path reveals an extensive network of uracil-specific interactions spanning the first 12 nucleotides of an oligoU-tailed RNA. We identify three U-specificity zones that explain how Dis3l2 recognizes, binds and processes uridylated pre-let-7 in the final step of the Lin28-let-7 pathway.


Subject(s)
Exoribonucleases/chemistry , Exoribonucleases/metabolism , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Animals , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Exosome Multienzyme Ribonuclease Complex/chemistry , Mice , MicroRNAs/chemistry , MicroRNAs/genetics , Models, Molecular , Oligoribonucleotides/chemistry , Oligoribonucleotides/metabolism , RNA-Binding Proteins/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Substrate Specificity , Uracil Nucleotides/chemistry , Uracil Nucleotides/metabolism
3.
RNA ; 23(10): 1582-1591, 2017 10.
Article in English | MEDLINE | ID: mdl-28698239

ABSTRACT

Arrays of singly labeled short oligonucleotides that hybridize to a specific target revolutionized RNA biology, enabling quantitative, single-molecule microscopy analysis and high-efficiency RNA/RNP capture. Here, we describe a simple and efficient method that allows flexible functionalization of inexpensive DNA oligonucleotides by different fluorescent dyes or biotin using terminal deoxynucleotidyl transferase and custom-made functional group conjugated dideoxy-UTP. We show that (i) all steps of the oligonucleotide labeling-including conjugation, enzymatic synthesis, and product purification-can be performed in a standard biology laboratory, (ii) the process yields >90%, often >95% labeled product with minimal carryover of impurities, and (iii) the oligonucleotides can be labeled with different dyes or biotin, allowing single-molecule FISH, RNA affinity purification, and Northern blot analysis to be performed.


Subject(s)
DNA Nucleotidylexotransferase/metabolism , In Situ Hybridization, Fluorescence/methods , RNA Probes/chemistry , Animals , Biotin , Dideoxynucleotides/chemistry , Dideoxynucleotides/metabolism , Drosophila melanogaster/genetics , Female , Fluorescent Dyes/chemistry , Oligonucleotide Probes/chemistry , Oligonucleotides/chemistry , Ovary/physiology , RNA Probes/metabolism , Uracil Nucleotides/chemistry , Uracil Nucleotides/metabolism
4.
Molecules ; 24(19)2019 Sep 25.
Article in English | MEDLINE | ID: mdl-31557948

ABSTRACT

Several health benefits, associated with human milk oligosaccharides (HMOS), have been revealed in the last decades. Further progress, however, requires not only the establishment of a simple "routine" method for absolute quantification of complex HMOS mixtures but also the development of novel synthesis strategies to improve access to tailored HMOS. Here, we introduce a combination of salvage-like nucleotide sugar-producing enzyme cascades with Leloir-glycosyltransferases in a sequential pattern for the convenient tailoring of stable isotope-labeled HMOS. We demonstrate the assembly of [13C6]galactose into lacto-N- and lacto-N-neo-type HMOS structures up to octaoses. Further, we present the enzymatic production of UDP-[15N]GlcNAc and its application for the enzymatic synthesis of [13C6/15N]lacto-N-neo-tetraose for the first time. An exemplary application was selected-analysis of tetraose in complex biological mixtures-to show the potential of tailored stable isotope reference standards for the mass spectrometry-based quantification, using matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF-MS) as a fast and straightforward method for absolute quantification of HMOS. Together with the newly available well-defined tailored isotopic HMOS, this can make a crucial contribution to prospective research aiming for a more profound understanding of HMOS structure-function relations.


Subject(s)
Milk, Human/chemistry , Oligosaccharides/chemistry , Carbon Isotopes/chemistry , Catalysis , Glycosyltransferases/chemistry , Humans , Nitrogen Isotopes/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Uracil Nucleotides/chemistry
5.
Molecules ; 24(23)2019 Nov 27.
Article in English | MEDLINE | ID: mdl-31783537

ABSTRACT

Dinucleoside 5',5'-polyphosphates (DNPs) are endogenous substances that play important intra- and extracellular roles in various biological processes, such as cell proliferation, regulation of enzymes, neurotransmission, platelet disaggregation and modulation of vascular tone. Various methodologies have been developed over the past fifty years to access these compounds, involving enzymatic processes or chemical procedures based either on P(III) or P(V) chemistry. Both solution-phase and solid-support strategies have been developed and are reported here. Recently, green chemistry approaches have emerged, offering attracting alternatives. This review outlines the main synthetic pathways for the preparation of dinucleoside 5',5'-polyphosphates, focusing on pharmacologically relevant compounds, and highlighting recent advances.


Subject(s)
Dinucleoside Phosphates/chemical synthesis , Purinergic P2Y Receptor Agonists/chemical synthesis , Deoxycytosine Nucleotides/agonists , Deoxycytosine Nucleotides/chemistry , Deoxycytosine Nucleotides/pharmacology , Dinucleoside Phosphates/chemistry , Dinucleoside Phosphates/isolation & purification , Dry Eye Syndromes/drug therapy , Green Chemistry Technology , Humans , Ophthalmic Solutions , Phosphorylation , Polyphosphates/chemical synthesis , Polyphosphates/chemistry , Purinergic P2Y Receptor Agonists/chemistry , Purinergic P2Y Receptor Agonists/isolation & purification , Receptors, Purinergic/metabolism , Uracil Nucleotides/chemistry , Uridine/agonists , Uridine/analogs & derivatives , Uridine/chemistry , Uridine/pharmacology
6.
Bioorg Med Chem Lett ; 28(7): 1248-1251, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29506959

ABSTRACT

A conjugate of triphosphorylated 2',3'-dideoxyuridine (ddU) with SiO2 nanoparticles was obtained via the CuAAC click chemistry between a γ-alkynyl ddU triphosphate and azido-modified SiO2 nanoparticles. Assessment of cytotoxicity in human breast adenocarcinoma MCF7 cells demonstrated that ddU triphosphate conjugated to SiO2 nanoparticles exhibited a 50% decrease in cancer cell growth at a concentration of 183 ±â€¯57 µg/mL, which corresponds to 22 ±â€¯7 µM of the parent nucleotide, whereas the parent nucleoside, nucleotide and alkynyl triphosphate precursor do not show any cytotoxicity. The data provide an example of remarkable potential of novel conjugates of SiO2 nanoparticles with phosphorylated nucleoside analogues, even those, which have not been used previously as therapeutics, for application as new anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Dideoxynucleotides/pharmacology , Nanoparticles/chemistry , Silicon Dioxide/pharmacology , Uracil Nucleotides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dideoxynucleotides/chemical synthesis , Dideoxynucleotides/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Molecular Structure , Silicon Dioxide/chemistry , Structure-Activity Relationship , Uracil Nucleotides/chemical synthesis , Uracil Nucleotides/chemistry
7.
Mol Cell ; 34(6): 710-21, 2009 Jun 26.
Article in English | MEDLINE | ID: mdl-19560423

ABSTRACT

We show that RNA polymerase (Pol) II prevents erroneous transcription in vitro with different strategies that depend on the type of DNARNA base mismatch. Certain mismatches are efficiently formed but impair RNA extension. Other mismatches allow for RNA extension but are inefficiently formed and efficiently proofread by RNA cleavage. X-ray analysis reveals that a TU mismatch impairs RNA extension by forming a wobble base pair at the Pol II active center that dissociates the catalytic metal ion and misaligns the RNA 3' end. The mismatch can also stabilize a paused state of Pol II with a frayed RNA 3' nucleotide. The frayed nucleotide binds in the Pol II pore either parallel or perpendicular to the DNA-RNA hybrid axis (fraying sites I and II, respectively) and overlaps the nucleoside triphosphate (NTP) site, explaining how it halts transcription during proofreading, before backtracking and RNA cleavage.


Subject(s)
Base Pair Mismatch , RNA Polymerase II/physiology , Transcription, Genetic/physiology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Structure, Tertiary , RNA Polymerase II/chemistry , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Thymine Nucleotides/chemistry , Thymine Nucleotides/metabolism , Uracil Nucleotides/chemistry , Uracil Nucleotides/metabolism
8.
Bioorg Med Chem ; 23(17): 5764-73, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26233801

ABSTRACT

P2Y6 receptor (P2Y6-R) is involved in various physiological and pathophysiological events. With a view to set rules for the design of UDP-based reversible P2Y6-R antagonists as potential drugs, we established structure-activity relationship of UDP analogues, bearing modifications at the uracil ring, ribose moiety, and the phosphate chain. For instance, C5-phenyl- or 3-NMe-uridine-5'-α,ß-methylene-diphosphonate, 16 and 23, or lack of 2'-OH, in 12-15, resulted in loss of both agonist and antagonist activity toward hP2Y6-R. However, uridylyl phosphosulfate, 19, selectively inhibited hP2Y6-R (IC50 112 µM) versus P2Y2/4-Rs. In summary, we have established a comprehensive SAR for hP2Y6-R ligands towards the development of hP2Y6-R antagonists.


Subject(s)
Receptors, Purinergic P2/chemistry , Uracil Nucleotides/chemical synthesis , Humans , Molecular Structure , Structure-Activity Relationship , Uracil Nucleotides/chemistry
9.
J Org Chem ; 79(11): 5315-9, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24819695

ABSTRACT

Jakeman et al. recently reported the inability to distinguish the diastereomers of uridine 5'-ß,γ-fluoromethylenetriphosphate (ß,γ-CHF-UTP, 1) by (19)F NMR under conditions we previously prescribed for the resolution of the corresponding ß,γ-CHF-dGTP spectra, stating further that 1 decomposed under these basic conditions. Here we show that the (19)F NMR spectra of 1 (~1:1 diastereomer mixture prepared by coupling of UMP-morpholidate with fluoromethylenebis(phosphonic acid)) in D2O at pH 10 are indeed readily distinguishable. 1 in this solution was stable for 24 h at rt.


Subject(s)
Deoxyguanine Nucleotides/chemistry , Fluorine/chemistry , Uracil Nucleotides/chemistry , Uridine/chemistry , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Models, Molecular , Stereoisomerism
10.
Int J Pharm ; 659: 124249, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38772496

ABSTRACT

This study aimed to develop, optimize, and evaluate hot-melt-extruded ophthalmic inserts capable of sustained release of diquafosol tetrasodium (DQS) via a design of experiments approach. DQS, a tear stimulant for dry eye management, faces challenges of frequent administration and low bioavailability. The developed insert uses biodegradable polymers in varied proportions to achieve sustained release. Optimized through mixture design, the insert completely dissolved within 24 h and maintained a stable drug content, thickness, and surface pH over three months at room temperature. In vitro corneal permeation studies on excised rabbit corneas demonstrated increased bioavailability, suggesting a reduced dosing frequency compared with conventional eye drops. Therefore, this insert has potential to enhance treatment outcomes by improving patient compliance and providing sustained drug effects.


Subject(s)
Cornea , Delayed-Action Preparations , Polyphosphates , Uracil Nucleotides , Rabbits , Animals , Polyphosphates/chemistry , Uracil Nucleotides/administration & dosage , Uracil Nucleotides/chemistry , Cornea/metabolism , Cornea/drug effects , Ophthalmic Solutions/administration & dosage , Ophthalmic Solutions/chemistry , Biological Availability , Drug Liberation , Administration, Ophthalmic , Drug Compounding/methods , Drug Implants , Hot Temperature , Chemistry, Pharmaceutical/methods
11.
ACS Appl Mater Interfaces ; 16(27): 34757-34771, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38946068

ABSTRACT

Dry eye disease (DED) is a chronic multifactorial ocular surface disease mainly caused by the instability of tear film, characterized by a series of ocular discomforts and even visual disorders. Oxidative stress has been recognized as an upstream factor in DED development. Diquafosol sodium (DQS) is an agonist of the P2Y2 receptor to restore the integrity/stability of the tear film. With the ability to alternate between Ce3+ and Ce4+, cerium oxide nanozymes could scavenge overexpressed reactive oxygen species (ROS). Hence, a DQS-loaded cerium oxide nanozyme was designed to boost the synergistic treatment of DED. Cerium oxide with branched polyethylenimine-graft-poly(ethylene glycol) as nucleating agent and dispersant was fabricated followed with DQS immobilization via a dynamic phenylborate ester bond, obtaining the DQS-loaded cerium oxide nanozyme (defined as Ce@PBD). Because of the ability to mimic the cascade processes of superoxide dismutase and catalase, Ce@PBD could scavenge excessive accumulated ROS, showing strong antioxidant and anti-inflammatory properties. Meanwhile, the P2Y2 receptors in the conjunctival cells could be stimulated by DQS in Ce@PBD, which can relieve the incompleteness and instability of the tear film. The animal experiments demonstrated that Ce@PBD significantly restored the defect of the corneal epithelium and increased the number of goblet cells, with the promotion of tear secretion, which was the best among commercial DQS ophthalmic solutions.


Subject(s)
Cerium , Dry Eye Syndromes , Cerium/chemistry , Cerium/pharmacology , Animals , Dry Eye Syndromes/drug therapy , Dry Eye Syndromes/pathology , Dry Eye Syndromes/metabolism , Uracil Nucleotides/chemistry , Uracil Nucleotides/pharmacology , Reactive Oxygen Species/metabolism , Humans , Antioxidants/chemistry , Antioxidants/pharmacology , Oxidative Stress/drug effects , Polyphosphates/chemistry , Polyphosphates/pharmacology , Mice , Rabbits
12.
Chembiochem ; 14(16): 2144-52, 2013 Nov 04.
Article in English | MEDLINE | ID: mdl-24106095

ABSTRACT

5-(hydroxymethyl)cytosine (5-hmC) is a newly identified oxidative product of 5-methylcytosine (5-mC) in the mammalian genome, and is believed to be an important epigenetic marker influencing a variety of biological processes. In addition to its relatively low abundance, the fluctuation of 5-hmC levels over time during cell development poses a formidable challenge for its accurate mapping and quantification. Here we describe a specific chemoenzymatic approach to 5-hmC detection in DNA samples by using new uridine 5'-diphosphoglucosamine (UDP-GlcN) probes. Our approach requires modification of the glucose moiety of UDP-Glc with small amino groups and transfer of these glucose derivatives to the hydroxy moiety of 5-hmC by using T4 phage glucosyltransferases. We evaluated the transfer efficiencies of three glucosyltransferases (wild-type α- and ß-GTs and a Y261L mutant ß-GT) with five different UDP-Glc derivatives containing functionalized groups for subsequent bioconjugation and detection. Our results indicate that UDP-6-N3 -Glc, UDP-6-GlcN, and UDP-2-GlcN can be transferred by ß-GT with efficiencies similar to that seen with the native UDP-Glc cofactor. 6-N3 -Glc- and 6-GlcN-containing oligonucleotides were selectively labeled with reactive fluorescent probes. In addition, a 2 kb DNA fragment modified with 2-GlcN groups was specifically detected by use of a commercially available antiglucosamine antibody. Alternative substrates for ß-GT and correlated glycosyltransferases might prove useful for the study of the function and dynamics of 5-hmC and other modified nucleotides, as well as for multiplex analysis.


Subject(s)
Cytosine/analogs & derivatives , Glucosamine/chemistry , Staining and Labeling , Uracil Nucleotides/chemistry , 5-Methylcytosine/analogs & derivatives , Animals , Antibodies/immunology , Bacteriophage T4/enzymology , Cytosine/chemistry , DNA/chemistry , DNA/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fluorescent Dyes/chemistry , Glucosamine/immunology , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycosylation , Mice , Oligonucleotides/chemical synthesis , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Substrate Specificity
13.
Org Biomol Chem ; 11(37): 6357-71, 2013 Oct 07.
Article in English | MEDLINE | ID: mdl-23945704

ABSTRACT

Derivatives of UMP (uridine monophosphate) with a fluorogenic substituent in position 5 represent a small but unique class of fluorophores, which has found important applications in chemical biology and biomolecular chemistry. In this study, we have synthesised a series of derivatives of the uracil nucleotides UMP, UDP and UTP with different aromatic and heteroaromatic substituents in position 5, in order to systematically investigate the influence of the 5-substituent on fluorescence emission. We have determined relevant photophysical parameters for all derivatives in this series, including quantum yields for the best fluorophores. The strongest fluorescence emission was observed with a 5-formylthien-2-yl substituent in position 5 of the uracil base, while the corresponding 3-formylthien-2-yl-substituted regioisomer was significantly less fluorescent. The 5-(5-formylthien-2-yl) uracil fluorophore was studied further in solvents of different polarity and proticity. In conjunction with results from a conformational analysis based on NMR data and computational experiments, these findings provide insights into the steric and electronic factors that govern fluorescence emission in this class of fluorophores. In particular, they highlight the interplay between fluorescence emission and conformation in this series. Finally, we carried out ligand-binding experiments with the 5-(5-formylthien-2-yl) uracil fluorophore and a UDP-sugar-dependent glycosyltransferase, demonstrating its utility for biological applications. The results from our photophysical and biological studies suggest, for the first time, a structural explanation for the fluorescence quenching effect that is observed upon binding of these fluorophores to a target protein.


Subject(s)
Fluorescent Dyes/chemistry , Uracil Nucleotides/chemistry , Computer Simulation , Molecular Conformation , Molecular Structure , Spectrometry, Fluorescence , Time Factors , Uracil Nucleotides/chemical synthesis
14.
Org Biomol Chem ; 10(33): 6785-91, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22825538

ABSTRACT

The general acid/base catalyzed cleavage of a number of alkyl esters of uridine-3'- (and -5'-)phosphate has been studied by utilizing a cleaving agent, in which the catalytic moiety (a substituted 1,3,5-triazine) is tethered to an anchoring Zn(II):cyclen moiety. Around pH 7, formation of a strong ternary complex between uracil, Zn(II) and cyclen brings the general acid/base catalyst close to the scissile phosphodiester linkage, resulting in rate acceleration of 1-2 orders of magnitude with the uridine-3'-phosphodiesters. Curiously, no acceleration was observed with their 5'-counterparts. A ß(lg) value of -0.7 has been determined for the general acid/base catalyzed cleavage, consistent with a proton transfer to the leaving group in the rate-limiting step.


Subject(s)
Heterocyclic Compounds/chemistry , RNA/chemistry , Uracil Nucleotides/chemistry , Zinc/chemistry , Acids/chemistry , Catalysis , Cyclams , Esters , Hydrogen-Ion Concentration , Triazines/chemistry
15.
Bioorg Med Chem ; 20(7): 2304-15, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22386981

ABSTRACT

We explored the influence of modifications of uridine 5'-methylenephosphonate on biological activity at the human P2Y(2) receptor. Key steps in the synthesis of a series of 5-substituted uridine 5'-methylenephosphonates were the reaction of a suitably protected uridine 5'-aldehyde with [(diethoxyphosphinyl)methylidene]triphenylphosphorane, C-5 bromination and a Suzuki-Miyaura coupling. These analogues behaved as selective agonists at the P2Y(2) receptor, with three analogues exhibiting potencies in the submicromolar range. Although maximal activities observed with the phosphonate analogues were much less than observed with UTP, high concentrations of the phosphonates had no effect on the stimulatory effect of UTP. These results suggest that these phosphonates bind to an allosteric site of the P2Y(2) receptor.


Subject(s)
Organophosphonates/chemistry , Purinergic P2Y Receptor Agonists/chemical synthesis , Receptors, Purinergic P2Y2/chemistry , Cell Line , Cell Proliferation/drug effects , Humans , Organophosphonates/chemical synthesis , Organophosphonates/pharmacology , Purinergic P2Y Receptor Agonists/chemistry , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y2/metabolism , Uracil Nucleotides/chemistry , Uridine Triphosphate/metabolism
16.
Nat Med ; 11(2): 223-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15665829

ABSTRACT

Beyond serving as a mechanical barrier, the endothelium has important regulatory functions. The discovery of nitric oxide revolutionized our understanding of vasoregulation. In contrast, the identity of endothelium-derived vasoconstrictive factors (EDCFs) remains unclear. The supernatant obtained from mechanically stimulated human endothelial cells obtained from dermal vessels elicited a vasoconstrictive response in an isolated perfused rat kidney. A purinoceptor blocker had a greater effect than an endothelin receptor blocker in decreasing endothelially derived vasoconstriction in the isolated perfused rat kidney. The nucleotide uridine adenosine tetraphosphate (Up(4)A) was isolated from the supernatant of stimulated human endothelium and identified by mass spectrometry. Up(4)A is likely to exert vasoconstriction predominantly through P2X1 receptors, and probably also through P2Y2 and P2Y4 receptors. Plasma concentrations of Up(4)A that cause vasoconstriction are found in healthy subjects. Stimulation with adenosine 5'-triphosphate (ATP), uridine 5'-triphosphate (UTP), acetylcholine, endothelin, A23187 and mechanical stress releases Up(4)A from endothelium, suggesting that Up(4)A contributes to vascular autoregulation. To our knowledge, Up(4)A is the first dinucleotide isolated from living organisms that contains both purine and pyrimidine moieties. We conclude that Up(4)A is a novel potent nonpeptidic EDCF. Its vasoactive effects, plasma concentrations and its release upon endothelial stimulation strongly suggest that Up(4)A has a functional vasoregulatory role.


Subject(s)
Adenine Nucleotides/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Uracil Nucleotides/metabolism , Vasoconstrictor Agents/metabolism , Adenine Nucleotides/chemistry , Animals , Cells, Cultured , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Humans , In Vitro Techniques , Kidney/cytology , Kidney/metabolism , Mass Spectrometry , Molecular Weight , Rats , Uracil Nucleotides/chemistry , Vasoconstrictor Agents/chemistry
17.
Nucleic Acids Res ; 38(14): 4868-76, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20223772

ABSTRACT

The side-by-side interactions of nucleobases contribute to the organization of RNA, forming the planar building blocks of helices and mediating chain folding. Dinucleotide platforms, formed by side-by-side pairing of adjacent bases, frequently anchor helices against loops. Surprisingly, GpU steps account for over half of the dinucleotide platforms observed in RNA-containing structures. Why GpU should stand out from other dinucleotides in this respect is not clear from the single well-characterized H-bond found between the guanine N2 and the uracil O4 groups. Here, we describe how an RNA-specific H-bond between O2'(G) and O2P(U) adds to the stability of the GpU platform. Moreover, we show how this pair of oxygen atoms forms an out-of-plane backbone 'edge' that is specifically recognized by a non-adjacent guanine in over 90% of the cases, leading to the formation of an asymmetric miniduplex consisting of 'complementary' GpUpA and GpA subunits. Together, these five nucleotides constitute the conserved core of the well-known loop-E motif. The backbone-mediated intrinsic stabilities of the GpU dinucleotide platform and the GpUpA/GpA miniduplex plausibly underlie observed evolutionary constraints on base identity. We propose that they may also provide a reason for the extreme conservation of GpU observed at most 5'-splice sites.


Subject(s)
Adenine Nucleotides/chemistry , Guanine Nucleotides/chemistry , RNA/chemistry , Uracil Nucleotides/chemistry , Evolution, Molecular , Hydrogen Bonding , Models, Molecular , RNA Stability
18.
Structure ; 16(1): 82-92, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18184586

ABSTRACT

UMP synthase (UMPS) catalyzes the last two steps of de novo pyrimidine nucleotide synthesis and is a potential cancer drug target. The C-terminal domain of UMPS is orotidine-5'-monophosphate decarboxylase (OMPD), a cofactor-less yet extremely efficient enzyme. Studies of OMPDs from micro-organisms led to the proposal of several noncovalent decarboxylation mechanisms via high-energy intermediates. We describe nine crystal structures of human OMPD in complex with substrate, product, and nucleotide inhibitors. Unexpectedly, simple compounds can replace the natural nucleotides and induce a closed conformation of OMPD, defining a tripartite catalytic site. The structures outline the requirements drugs must meet to maximize therapeutic effects and minimize cross-species activity. Chemical mimicry by iodide identified a CO(2) product binding site. Plasticity of catalytic residues and a covalent OMPD-UMP complex prompt a reevaluation of the prevailing decarboxylation mechanism in favor of covalent intermediates. This mechanism can also explain the observed catalytic promiscuity of OMPD.


Subject(s)
Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Orotate Phosphoribosyltransferase/chemistry , Orotate Phosphoribosyltransferase/metabolism , Orotidine-5'-Phosphate Decarboxylase/chemistry , Orotidine-5'-Phosphate Decarboxylase/metabolism , Drug Design , Humans , Kinetics , Models, Molecular , Orotidine-5'-Phosphate Decarboxylase/drug effects , Protein Conformation , Uracil Nucleotides/chemistry , Uracil Nucleotides/metabolism
19.
Bioorg Med Chem ; 17(14): 5298-311, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19502066

ABSTRACT

The P2Y(14) receptor, a nucleotide signaling protein, is activated by uridine-5'-diphosphoglucose 1 and other uracil nucleotides. We have determined that the glucose moiety of 1 is the most structurally permissive region for designing analogues of this P2Y(14) agonist. For example, the carboxylate group of uridine-5'-diphosphoglucuronic acid proved to be suitable for flexible substitution by chain extension through an amide linkage. Functionalized congeners containing terminal 2-acylaminoethylamides prepared by this strategy retained P2Y(14) activity, and molecular modeling predicted close proximity of this chain to the second extracellular loop of the receptor. In addition, replacement of glucose with other sugars did not diminish P2Y(14) potency. For example, the [5'']ribose derivative had an EC(50) of 0.24muM. Selective monofluorination of the glucose moiety indicated a role for the 2''- and 6''-hydroxyl groups of 1 in receptor recognition. The beta-glucoside was twofold less potent than the native alpha-isomer, but methylene replacement of the 1''-oxygen abolished activity. Replacement of the ribose ring system with cyclopentyl or rigid bicyclo[3.1.0]hexane groups abolished activity. Uridine-5'-diphosphoglucose also activates the P2Y(2) receptor, but the 2-thio analogue and several of the potent modified-glucose analogues were P2Y(14)-selective.


Subject(s)
Purinergic P2 Receptor Agonists , Receptors, Purinergic P2/metabolism , Structure-Activity Relationship , Uracil Nucleotides/chemistry , Uracil Nucleotides/pharmacology , Uridine Diphosphate Glucose/analogs & derivatives , Animals , COS Cells , Chlorocebus aethiops , Humans , Models, Molecular , Molecular Structure , Protein Binding , Protein Conformation , Receptors, Purinergic P2/chemistry , Type C Phospholipases/metabolism , Uracil Nucleotides/chemical synthesis
20.
Nat Commun ; 10(1): 5292, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31757970

ABSTRACT

Message RNA poly(A) tails are vital for their function and regulation. However, the full-length sequence of mRNA isoforms with their poly(A) tails remains undetermined. Here, we develop a method at single-cell level sensitivity that enables quantification of poly(A) tails along with the full-length cDNA while reading non-adenosine residues within poly(A) tails precisely, which we name poly(A) inclusive RNA isoform sequencing (PAIso-seq). Using this method, we can quantify isoform specific poly(A) tail length. More interestingly, we find that 17% of the mRNAs harbor non-A residues within the body of poly(A) tails in mouse GV oocytes. We show that PAIso-seq is sensitive enough to analyze single GV oocytes. These findings will not only provide an accurate and sensitive tool in studying poly(A) tails, but also open a door for the function and regulation of non-adenosine modifications within the body of poly(A) tails.


Subject(s)
Cytosine Nucleotides/chemistry , Guanine Nucleotides/chemistry , Oocytes/metabolism , Poly A/chemistry , RNA, Messenger/chemistry , Sequence Analysis, RNA/methods , Uracil Nucleotides/chemistry , Animals , Cell Cycle Proteins/genetics , Cyclin B1/genetics , Cytosine Nucleotides/analysis , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Guanine Nucleotides/analysis , Mice , Poly A/analysis , Protein Biosynthesis , RNA Isoforms , RNA, Messenger/analysis , Single-Cell Analysis , Tissue Plasminogen Activator/genetics , Uracil Nucleotides/analysis
SELECTION OF CITATIONS
SEARCH DETAIL