Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.479
Filter
1.
Stem Cell Res Ther ; 14(1): 97, 2023 04 19.
Article in English | MEDLINE | ID: mdl-37076930

ABSTRACT

BACKGROUND: Endogenously released adenine and uracil nucleotides favour the osteogenic commitment of bone marrow-derived mesenchymal stromal cells (BM-MSCs) through the activation of ATP-sensitive P2X7 and UDP-sensitive P2Y6 receptors. Yet, these nucleotides have their osteogenic potential compromised in post-menopausal (Pm) women due to overexpression of nucleotide metabolizing enzymes, namely NTPDase3. This prompted us to investigate whether NTPDase3 gene silencing or inhibition of its enzymatic activity could rehabilitate the osteogenic potential of Pm BM-MSCs. METHODS: MSCs were harvested from the bone marrow of Pm women (69 ± 2 years old) and younger female controls (22 ± 4 years old). The cells were allowed to grow for 35 days in an osteogenic-inducing medium in either the absence or the presence of NTPDase3 inhibitors (PSB 06126 and hN3-B3s antibody); pre-treatment with a lentiviral short hairpin RNA (Lenti-shRNA) was used to silence the NTPDase3 gene expression. Immunofluorescence confocal microscopy was used to monitor protein cell densities. The osteogenic commitment of BM-MSCs was assessed by increases in the alkaline phosphatase (ALP) activity. The amount of the osteogenic transcription factor Osterix and the alizarin red-stained bone nodule formation. ATP was measured with the luciferin-luciferase bioluminescence assay. The kinetics of the extracellular ATP (100 µM) and UDP (100 µM) catabolism was assessed by HPLC RESULTS: The extracellular catabolism of ATP and UDP was faster in BM-MSCs from Pm women compared to younger females. The immunoreactivity against NTPDase3 increased 5.6-fold in BM-MSCs from Pm women vs. younger females. Selective inhibition or transient NTPDase3 gene silencing increased the extracellular accumulation of adenine and uracil nucleotides in cultured Pm BM-MSCs. Downregulation of NTPDase3 expression or activity rehabilitated the osteogenic commitment of Pm BM-MSCs measured as increases in ALP activity, Osterix protein cellular content and bone nodule formation; blockage of P2X7 and P2Y6 purinoceptors prevented this effect. CONCLUSIONS: Data suggest that NTPDase3 overexpression in BM-MSCs may be a clinical surrogate of the osteogenic differentiation impairment in Pm women. Thus, besides P2X7 and P2Y6 receptors activation, targeting NTPDase3 may represent a novel therapeutic strategy to increase bone mass and reduce the osteoporotic risk of fractures in Pm women.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Humans , Female , Aged , Adolescent , Young Adult , Adult , Postmenopause , Mesenchymal Stem Cells/metabolism , Cell Differentiation , Uracil Nucleotides/metabolism , Uracil Nucleotides/pharmacology , Uridine Diphosphate/metabolism , Uridine Diphosphate/pharmacology , Adenosine Triphosphate/metabolism , Bone Marrow Cells , Cells, Cultured
2.
Curr Opin Pharmacol ; 63: 102190, 2022 04.
Article in English | MEDLINE | ID: mdl-35231671

ABSTRACT

Extracellular uridine nucleotides regulate physiological and pathophysiological metabolic processes through the activation of P2Y2, P2Y4, P2Y6 and P2Y14 purinergic receptors, which play a key role in adipogenesis, glucose uptake, lipolysis and adipokine secretion. Using adipocyte-specific knockout mouse models, it has been demonstrated that lack of the P2Y6R or P2Y14R can protect against diet-induced obesity and improve whole-body glucose metabolism. The P2Y2R facilitated adipogenesis and inflammation, and the loss of P2Y4R or P2Y14R raised the levels of the protective endocrine factor adiponectin. Hence, potent antagonists for these receptors may be tested to identify drug candidates for the treatment of obesity and type 2 diabetes. However, future studies are required to provide insight into purinergic regulation of brown adipocytes and their role in thermogenesis. This review summarizes the current studies on uridine nucleotide-activated P2YRs and their role in adipocyte function, diet-induced obesity and associated metabolic deficits.


Subject(s)
Diabetes Mellitus, Type 2 , Uracil Nucleotides , Adipocytes/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Humans , Mice , Obesity/drug therapy , Obesity/metabolism , Receptors, Purinergic/metabolism , Uracil Nucleotides/metabolism , Uracil Nucleotides/pharmacology
3.
Science ; 375(6577): 161-167, 2022 Jan 14.
Article in English | MEDLINE | ID: mdl-34855509

ABSTRACT

The COVID-19 pandemic has underscored the critical need for broad-spectrum therapeutics against respiratory viruses. Respiratory syncytial virus (RSV) is a major threat to pediatric patients and older adults. We describe 4'-fluorouridine (4'-FlU, EIDD-2749), a ribonucleoside analog that inhibits RSV, related RNA viruses, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with high selectivity index in cells and human airway epithelia organoids. Polymerase inhibition within in vitro RNA-dependent RNA polymerase assays established for RSV and SARS-CoV-2 revealed transcriptional stalling after incorporation. Once-daily oral treatment was highly efficacious at 5 milligrams per kilogram (mg/kg) in RSV-infected mice or 20 mg/kg in ferrets infected with different SARS-CoV-2 variants of concern, initiated 24 or 12 hours after infection, respectively. These properties define 4'-FlU as a broad-spectrum candidate for the treatment of RSV, SARS-CoV-2, and related RNA virus infections.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , SARS-CoV-2/drug effects , Uracil Nucleotides/pharmacology , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/metabolism , COVID-19/virology , Cell Line , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Disease Models, Animal , Female , Ferrets , Humans , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mononegavirales/drug effects , Mononegavirales/physiology , RNA-Dependent RNA Polymerase/metabolism , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/physiology , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Transcription, Genetic , Uracil Nucleotides/administration & dosage , Uracil Nucleotides/metabolism , Virus Replication/drug effects
4.
J Immunol ; 207(5): 1275-1287, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34389624

ABSTRACT

The airway epithelial cells (AECs) lining the conducting passageways of the lung secrete a variety of immunomodulatory factors. Among these, PGE2 limits lung inflammation and promotes bronchodilation. By contrast, IL-6 drives intense airway inflammation, remodeling, and fibrosis. The signaling that differentiates the production of these opposing mediators is not understood. In this study, we find that the production of PGE2 and IL-6 following stimulation of human AECs by the damage-associated molecular pattern extracellular ATP shares a common requirement for Ca2+ release-activated Ca2+ (CRAC) channels. ATP-mediated synthesis of PGE2 required activation of metabotropic P2Y2 receptors and CRAC channel-mediated cytosolic phospholipase A2 signaling. By contrast, ATP-evoked synthesis of IL-6 occurred via activation of ionotropic P2X receptors and CRAC channel-mediated calcineurin/NFAT signaling. In contrast to ATP, which elicited the production of both PGE2 and IL-6, the uridine nucleotide, UTP, stimulated PGE2 but not IL-6 production. These results reveal that human AECs employ unique receptor-specific signaling mechanisms with CRAC channels as a signaling nexus to regulate release of opposing immunomodulatory mediators. Collectively, our results identify P2Y2 receptors, CRAC channels, and P2X receptors as potential intervention targets for airway diseases.


Subject(s)
Dinoprostone/metabolism , Inflammation/immunology , Interleukin-6/metabolism , Respiratory Mucosa/metabolism , Adenosine Triphosphate/pharmacokinetics , Alarmins/metabolism , Calcium Release Activated Calcium Channels/metabolism , Cells, Cultured , Humans , Immunomodulation , Interleukin-6/genetics , NFATC Transcription Factors/metabolism , Phospholipases A2/metabolism , Receptors, Purinergic P2X/metabolism , Respiratory Mucosa/pathology , Signal Transduction , Uracil Nucleotides/metabolism
5.
Bioorg Med Chem Lett ; 30(23): 127539, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32919013

ABSTRACT

Nucleotide prodrugs are of great clinical interest for treating a variety of viral infections due to their ability to target tissues selectively and to deliver relatively high concentrations of the active nucleotide metabolite intracellularly. However, their clinical successes have been limited, oftentimes due to unwanted in vivo metabolic processes that reduce the quantities of nucleoside triphosphate that reach the site of action. In an attempt to circumvent this, we designed novel nucleosides that incorporate a sterically bulky group at the 5'-carbon of the phosphoester prodrug, which we reasoned would reduce the amounts of non-productive PO bond cleavage back to the corresponding nucleoside by nucleotidases. Molecular docking studies with the NS5B HCV polymerase suggested that a nucleotide containing a 5'-methyl group could be accommodated. Therefore, we synthesized mono- and diphosphate prodrugs of 2',5'-C-dimethyluridine stereoselectively and evaluated their cytotoxicity and anti-HCV activity in the HCV replicon assay. All four prodrugs exhibited anti-HCV activity with IC50 values in the single digit micromolar concentrations, with the 5'(R)-C-methyl prodrug displaying superior potency relative to its 5'(S)-C-methyl counterpart. However, when compared to the unmethylated prodrug, the potency is poorer. The poorer potency of these prodrugs may be due to unfavorable steric interactions of the 5'-C-methyl group in the active sites of the kinases that catalyze the formation of active triphosphate metabolite.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Prodrugs/pharmacology , Uracil Nucleotides/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Cell Line , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Prodrugs/chemical synthesis , Prodrugs/metabolism , Protein Binding , Uracil Nucleotides/chemical synthesis , Uracil Nucleotides/metabolism , Viral Nonstructural Proteins/metabolism
6.
Nat Commun ; 11(1): 2587, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32444616

ABSTRACT

The gut microbiota metabolizes drugs and alters their efficacy and toxicity. Diet alters drugs, the metabolism of the microbiota, and the host. However, whether diet-triggered metabolic changes in the microbiota can alter drug responses in the host has been largely unexplored. Here we show that dietary thymidine and serine enhance 5-fluoro 2'deoxyuridine (FUdR) toxicity in C. elegans through different microbial mechanisms. Thymidine promotes microbial conversion of the prodrug FUdR into toxic 5-fluorouridine-5'-monophosphate (FUMP), leading to enhanced host death associated with mitochondrial RNA and DNA depletion, and lethal activation of autophagy. By contrast, serine does not alter FUdR metabolism. Instead, serine alters E. coli's 1C-metabolism, reduces the provision of nucleotides to the host, and exacerbates DNA toxicity and host death without mitochondrial RNA or DNA depletion; moreover, autophagy promotes survival in this condition. This work implies that diet-microbe interactions can alter the host response to drugs without altering the drug or the host.


Subject(s)
Caenorhabditis elegans/drug effects , Floxuridine/toxicity , Food-Drug Interactions , Gastrointestinal Microbiome/drug effects , Serine/pharmacology , Animals , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Dietary Supplements , Escherichia coli/drug effects , Escherichia coli/metabolism , Floxuridine/pharmacokinetics , Folic Acid/metabolism , Gastrointestinal Microbiome/physiology , Thymidine/analogs & derivatives , Thymidine/metabolism , Thymidine/pharmacokinetics , Thymidine/pharmacology , Uracil Nucleotides/metabolism , Uracil Nucleotides/pharmacokinetics
7.
J Med Chem ; 62(9): 4555-4570, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30951311

ABSTRACT

We report the synthesis and biological evaluation of a series of 4'-fluoro-2'- C-substituted uridines. Triphosphates of the uridine analogues exhibited a potent inhibition of hepatitis C virus (HCV) NS5B polymerase with IC50 values as low as 27 nM. In an HCV subgenomic replicon assay, the phosphoramidate prodrugs of these uridine analogues demonstrated a very potent activity with EC50 values as low as 20 nM. A lead compound AL-335 (53) demonstrated high levels of the nucleoside triphosphate in vitro in primary human hepatocytes and Huh-7 cells as well as in dog liver following a single oral dose. Compound 53 was selected for the clinical development where it showed promising results in phase 1 and 2 trials.


Subject(s)
Alanine/analogs & derivatives , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Prodrugs/pharmacology , Uracil Nucleotides/pharmacology , Uridine/analogs & derivatives , Alanine/chemical synthesis , Alanine/pharmacology , Animals , Antiviral Agents/chemical synthesis , Cell Line, Tumor , Dogs , Hepacivirus/enzymology , Hepatitis C/drug therapy , Humans , Nucleic Acid Synthesis Inhibitors/chemical synthesis , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphoramides , Prodrugs/chemical synthesis , Replicon/drug effects , Uracil Nucleotides/chemical synthesis , Uracil Nucleotides/metabolism , Uridine/chemical synthesis , Uridine/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors
8.
Brain Res Bull ; 151: 12-24, 2019 09.
Article in English | MEDLINE | ID: mdl-30922852

ABSTRACT

P2Y receptors are G-protein-coupled receptors (GPCRs) for extracellular nucleotides. There are eight mammalian P2Y receptor subtypes divided into two subgroups (P2Y1, P2Y2, P2Y4, P2Y6, and P2Y11) and (P2Y12, P2Y13, and P2Y14). The P2Y receptors are expressed in various cell types and play important roles in physiology and pathophysiology including inflammatory responses and neuropathic pain. The antagonism of P2Y12 receptors is used in pharmacotherapy for the prevention and therapy of cardiovascular events. The nucleoside analogue ticagrelor and active metabolites of the thienopyridine compounds ticlopidine, clopidogrel and prasugrel inhibit platelet P2Y12 receptors and reduce thereby platelet aggregation. The P2Y2 receptor agonist diquafosol is used for the treatment of the dry eye syndrome. The P2Y receptor subtypes differ in their amino acid sequences, their pharmacological profiles and their signaling transduction pathways. Recently, selective receptor ligands have been developed for all subtypes. The published crystal structures of the human P2Y1 and P2Y12 receptors as well as receptor models will facilitate the development of novel drugs for pharmacotherapy.


Subject(s)
Receptors, Purinergic P2Y/metabolism , Receptors, Purinergic P2Y/physiology , Adenine Nucleotides/metabolism , Animals , Humans , Platelet Aggregation , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Receptors, Purinergic P2Y/ultrastructure , Signal Transduction , Structure-Activity Relationship , Uracil Nucleotides/metabolism
9.
J Med Chem ; 61(9): 3939-3951, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29681152

ABSTRACT

Ectonucleotide pyrophosphatase/phosphodiesterase-1 (NPP1) hydrolyzes phosphodiester bonds of nucleotides such as ATP, resulting mainly in the formation of AMP and pyrophosphate. NPP1 activity plays a deleterious function in calcified aortic valve disease and calcium pyrophosphate deposition disease. Thus, inhibitors of NPP1 represent a medical need. We developed novel NPP1 inhibitors based on uridine 5'-Pα,α-dithiophosphate analogues, 9-12. All these analogues potently inhibited hNPP1 (80-100% inhibition) at 100 µM, with no, or minimal, inhibition of NPP3 and other ectonucleotidases (NTPDase1,2,3,8). These compounds showed nearly no activity at uracil-nucleotide sensitive P2Y2,4,6-receptors and thus represent highly selective NPP1 inhibitors. The most promising inhibitor was diuridine 5'-Pα,α,5″-Pα,α-tetrathiotetraphosphate, 12, exhibiting Ki of 27 nM. Analogues 9-12 proved to be highly stable to air oxidation and to acidic and basic pH. Docking simulations suggested that the enhanced NPP1 inhibitory activity and selectivity of analogue 12 could be attributed to the simultaneous occupancy of two sites (the AMP site and an alternative site) of NPP1 by this compound.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Pyrophosphatases/antagonists & inhibitors , Uracil Nucleotides/chemistry , Uracil Nucleotides/pharmacology , Drug Stability , Enzyme Inhibitors/metabolism , Humans , Hydrolysis , Inhibitory Concentration 50 , Molecular Docking Simulation , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Protein Conformation , Pyrophosphatases/chemistry , Pyrophosphatases/metabolism , Structure-Activity Relationship , Substrate Specificity , Uracil Nucleotides/metabolism
10.
Pharmacol Ther ; 190: 24-80, 2018 10.
Article in English | MEDLINE | ID: mdl-29660366

ABSTRACT

P2Y receptors (P2YRs) are a family of G protein-coupled receptors activated by extracellular nucleotides. Physiological P2YR agonists include purine and pyrimidine nucleoside di- and triphosphates, such as ATP, ADP, UTP, UDP, nucleotide sugars, and dinucleotides. Eight subtypes exist, P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, and P2Y14, which represent current or potential future drug targets. Here we provide a comprehensive overview of ligands for the subgroup of the P2YR family that is activated by uracil nucleotides: P2Y2 (UTP, also ATP and dinucleotides), P2Y4 (UTP), P2Y6 (UDP), and P2Y14 (UDP, UDP-glucose, UDP-galactose). The physiological agonists are metabolically unstable due to their fast hydrolysis by ectonucleotidases. A number of agonists with increased potency, subtype-selectivity and/or enzymatic stability have been developed in recent years. Useful P2Y2R agonists include MRS2698 (6-01, highly selective) and PSB-1114 (6-05, increased metabolic stability). A potent and selective P2Y2R antagonist is AR-C118925 (10-01). For studies of the P2Y4R, MRS4062 (3-15) may be used as a selective agonist, while PSB-16133 (10-06) is a selective antagonist. Several potent P2Y6R agonists have been developed including 5-methoxyuridine 5'-O-((Rp)α-boranodiphosphate) (6-12), PSB-0474 (3-11), and MRS2693 (3-26). The isocyanate MRS2578 (10-08) is used as a selective P2Y6R antagonist, although its reactivity and low water-solubility are limiting. With MRS2905 (6-08), a potent and metabolically stable P2Y14R agonist is available, while PPTN (10-14) represents a potent and selective P2Y14R antagonist. The radioligand [3H]UDP can be used to label P2Y14Rs. In addition, several fluorescent probes have been developed. Uracil nucleotide-activated P2YRs show great potential as drug targets, especially in inflammation, cancer, cardiovascular and neurodegenerative diseases.


Subject(s)
Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Animals , Drug Development/methods , Humans , Ligands , Purinergic P2Y Receptor Antagonists/chemistry , Radioligand Assay , Receptors, Purinergic P2Y/metabolism , Solubility , Uracil Nucleotides/metabolism
11.
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
12.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 43-51, 2017 01.
Article in English | MEDLINE | ID: mdl-27768902

ABSTRACT

The Ubiquitin-Proteasome System (UPS) is essential for the regulation of the cellular proteostasis. Indeed, it has been postulated that an UPS dysregulation is the common mechanism that underlies several neurological disorders. Considering that extracellular nucleotides, through their selective P2Y2 receptor (P2Y2R), play a neuroprotective role in various neurological disorders that course with an UPS impairment, we wonder if this neuroprotective capacity resulted from their ability to modulate the UPS. Using a cellular model expressing two different UPS reporters, we found that the stimulation of P2Y2R by its selective agonist Up4U induced a significant reduction of UPS reporter levels. This reduction was due to an increase in two of the three peptidase proteasome activities, chymotrypsin and postglutamyl, caused by an increased expression of proteasome constitutive catalytic subunits ß1 and ß5. The intracellular signaling pathway involved required the activation of IP3/MEK1/2/ERK but was independent of PKC or PKA. Interestingly, the P2Y2R activation was able to revert both UPS-reporter accumulation and the cell death induced by a prolonged inhibition of UPS. Finally, we also observed that intracerebroventricular administration of Up4U induced a significant increase both of chymotrypsin and postglutamyl activities as well as an increased expression of proteasome subunits ß1 and ß5 in the hippocampus of wild-type mice, but not in P2Y2R KO mice. All these results strongly suggest that the capacity to modulate the UPS activity via P2Y2R is the molecular mechanism which is how the nucleotides play a neuroprotective role in neurological disorders.


Subject(s)
Neurons/drug effects , Neuroprotective Agents/pharmacology , Nucleotides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y2/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Inositol 1,4,5-Trisphosphate/metabolism , MAP Kinase Signaling System/drug effects , Mice , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/metabolism , Nucleotides/metabolism , Purinergic P2Y Receptor Agonists/metabolism , Uracil Nucleotides/metabolism , Uracil Nucleotides/pharmacology
13.
Plant J ; 88(5): 705-716, 2016 12.
Article in English | MEDLINE | ID: mdl-27490826

ABSTRACT

Despite the importance of riboflavin as the direct precursor of the cofactors flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN), the physiologically relevant catalyst dephosphorylating the riboflavin biosynthesis pathway intermediate 5-amino-6-ribitylamino-2,4(1H,3H) pyrimidinedione 5'-phosphate (ARPP) has not been characterized from any organism. By using as the query sequence a previously identified plastidial FMN hydrolase AtcpFHy1 (At1g79790), belonging to the haloacid dehalogenase (HAD) superfamily, seven candidates for the missing ARPP phosphatase were found, cloned, recombinantly expressed, and purified. Activity screening showed that the enzymes encoded by AtcpFHy1, At4g11570, and At4g25840 catalyze dephosphorylation of ARPP. AtcpFHy1 was renamed AtcpFHy/PyrP1, At4g11570 and At4g25840 were named AtPyrP2 and AtGpp1/PyrP3, respectively. Subcellular localization in planta indicated that AtPyrP2 was localized in plastids and AtGpp1/PyrP3 in mitochondria. Biochemical characterization of AtcpFHy/PyrP1 and AtPyrP2 showed that they have similar Km values for the substrate ARPP, with AtcpFHy/PyrP1 having higher catalytic efficiency. Screening of 21 phosphorylated substrates showed that AtPyrP2 is specific for ARPP. Molecular weights of AtcpFHy/PyrP1 and AtPyrP2 were estimated at 46 and 72 kDa, suggesting dimers. pH and temperature optima for AtcpFHy/PyrP1 and AtPyrP2 were ~7.0-8.5 and 40-50°C. T-DNA knockout of AtcpFHy/PyrP1 did not affect the flavin profile of the transgenic plants, whereas silencing of AtPyrP2 decreased accumulation of riboflavin, FMN, and FAD. Our results strongly support AtPyrP2 as the missing phosphatase on the riboflavin biosynthesis pathway in Arabidopsis thaliana. The identification of this enzyme closes a long-standing gap in understanding of the riboflavin biosynthesis in plants.


Subject(s)
Arabidopsis/enzymology , Arabidopsis/metabolism , Hydrolases/metabolism , Riboflavin/biosynthesis , Dinitrocresols/metabolism , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/metabolism , Uracil Nucleotides/metabolism
14.
Methods Mol Biol ; 1428: 93-114, 2016.
Article in English | MEDLINE | ID: mdl-27236794

ABSTRACT

Since DNA and histone levels must be closely balanced for cell survival, histone expressions are highly regulated. The regulation of replication-dependent histone expression is mainly achieved at the mRNA level, as the mRNAs are rapidly removed when DNA replication is inhibited during S-phase. Histone mRNA degradation initiates with addition of multiple uridines (oligouridylation) following the 3' stem-loop (SL) catalyzed by terminal uridyltransferase (TUTase). Previous studies showed that histone mRNA degradation occurs through both 5' → 3' and 3' → 5' processes, but the relative contributions are difficult to dissect due to lack of established protocols. The translational efficiency and stability of synthetic mRNA in both cultured cells and whole animals can be improved by structural modifications at the both 5' and 3' termini. In this chapter, we present methods of utilizing modified cap dinucleotide analogs to block 5' → 3' degradation of a reporter mRNA containing canonical histone mRNA 3' SL and monitoring how oligouridylation and 3' → 5' degradation occur. Protocols are presented for synthesis of reporter mRNA containing the histone 3' SL and modified cap analogs, monitoring mRNA stability and unidirectional degradation either from 5' or 3' termini, and detection of oligo(U) tracts from degradation products by either traditional or deep sequencing.


Subject(s)
Histones/genetics , RNA Cap Analogs/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , HeLa Cells , Humans , Oligoribonucleotides/metabolism , Protein Biosynthesis , RNA Nucleotidyltransferases/metabolism , RNA Stability , Synthetic Biology , Uracil Nucleotides/metabolism
15.
Mol Cell ; 61(3): 364-378, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26833087

ABSTRACT

Small, noncoding RNA biogenesis typically involves cleavage of structured precursor by RNase III-like endonucleases. However, guide RNAs (gRNAs) that direct U-insertion/deletion mRNA editing in mitochondria of trypanosomes maintain 5' triphosphate characteristic of the transcription initiation and possess a U-tail indicative of 3' processing and uridylation. Here, we identified a protein complex composed of RET1 TUTase, DSS1 3'-5' exonuclease, and three additional subunits. This complex, termed mitochondrial 3' processome (MPsome), is responsible for primary uridylation of ∼800 nt gRNA precursors, their processive degradation to a mature size of 40-60 nt, and secondary U-tail addition. Both strands of the gRNA gene are transcribed into sense and antisense precursors of similar lengths. Head-to-head hybridization of these transcripts blocks symmetrical 3'-5' degradation at a fixed distance from the double-stranded region. Together, our findings suggest a model in which gRNA is derived from the 5' extremity of a primary molecule by uridylation-induced, antisense transcription-controlled 3'-5' exonucleolytic degradation.


Subject(s)
Exoribonucleases/metabolism , Mitochondria/metabolism , RNA Editing , RNA, Antisense/metabolism , RNA, Guide, Kinetoplastida/biosynthesis , RNA, Protozoan/biosynthesis , RNA/biosynthesis , Trypanosoma brucei brucei/metabolism , Exoribonucleases/genetics , Gene Expression Regulation , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA/genetics , RNA Nucleotidyltransferases/genetics , RNA Nucleotidyltransferases/metabolism , RNA Stability , RNA, Antisense/genetics , RNA, Guide, Kinetoplastida/genetics , RNA, Mitochondrial , RNA, Protozoan/genetics , Time Factors , Trypanosoma brucei brucei/genetics , Uracil Nucleotides/metabolism
16.
Sci Rep ; 5: 16861, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26578061

ABSTRACT

Endogenous ribonucleotides and deoxyribonucleotides are essential metabolites that play important roles in a broad range of key cellular functions. Their intracellular levels could also reflect the action of nucleoside analogues. We investigated the effects of 5-fluorouracil (5-FU) on ribonucleotide and deoxyribonucleotide pool sizes in cells upon exposure to 5-FU for different durations. Unsupervised and supervised artificial neural networks were compared for comprehensive analysis of global responses to 5-FU. As expected, deoxyuridine monophosphate (dUMP) increased after 5-FU incubation due to the inhibition of thymine monophosphate (TMP) synthesis. Interestingly, the accumulation of dUMP could not lead to increased levels of deoxyuridine triphosphate (dUTP) and deoxyuridine diphosphate (dUDP). After the initial fall in intracellular deoxythymidine triphosphate (TTP) concentration, its level recovered and increased from 48 h exposure to 5-FU, although deoxythymidine diphosphate (TDP) and TMP continued to decrease compared with the control group. These findings suggest 5-FU treatment caused unexpected changes in intracellular purine polls, such as increases in deoxyadenosine triphosphate (dATP), adenosine-triphosphate (ATP), guanosine triphosphate (GTP) pools. Further elucidation of the mechanism of action of 5-FU in causing these changes should enhance development of strategies that will increase the anticancer activity of 5-FU while decreasing its resistance.


Subject(s)
Deoxyribonucleotides/metabolism , Fluorouracil/pharmacology , Metabolic Networks and Pathways/drug effects , Models, Biological , Neural Networks, Computer , Ribonucleotides/metabolism , Adenine Nucleotides/metabolism , Cell Survival/drug effects , Chromatography, Liquid , Cytosine Nucleotides/metabolism , Guanine Nucleotides , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Tandem Mass Spectrometry , Thymine Nucleotides/metabolism , Uracil Nucleotides/metabolism
17.
Carbohydr Res ; 414: 8-14, 2015 Sep 23.
Article in English | MEDLINE | ID: mdl-26162744

ABSTRACT

UDP-sugar 4-epimerase (GalE) belongs to the short-chain dehydrogenase/reductase (SDR) superfamily of proteins and is one of enzymes in the Leloir pathway. They have been shown to be important virulence factors in a number of Gram-negative pathogens and to be involved in the biosynthesis of different polysaccharide structures. The metabolic disease type III galactosemia is caused by detrimental mutations in the human GalE. GalE and related enzymes display unusual enzymologic, chemical, and stereochemical properties; including irreversible binding of the cofactor NAD and uridine nucleotide-induced activation of this cofactor. These epimerases have been found active on UDP-hexoses, the N-acetylated and uronic acid forms thereof as well as UDP-pentoses. As they are involved in different pathways and functions, a deeper understanding of the enzymes, and their substrate promiscuity and/or selectivity, could lead to drug and vaccine design as well as antibiotic and probiotic development. This review summarizes the research performed on UDP-sugar 4-epimerases' structure, mechanism and substrate promiscuity.


Subject(s)
Galactosemias/genetics , UDPglucose 4-Epimerase/chemistry , UDPglucose 4-Epimerase/metabolism , Crystallography, X-Ray , Galactosemias/enzymology , Humans , Models, Molecular , Mutation , NAD/metabolism , Protein Structure, Tertiary , Substrate Specificity , UDPglucose 4-Epimerase/genetics , Uracil Nucleotides/metabolism
18.
FEBS J ; 282(18): 3489-99, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26183531

ABSTRACT

RNA molecules are subjected to post-transcriptional modifications that might determine their maturation, activity, localization and stability. These alterations can occur within the RNA molecule or at its 5'- or 3'- extremities, and are essential for gene regulation and proper function of the RNA. One major type of modification is the 3'-end addition of nontemplated nucleotides. Polyadenylation is the most well studied type of 3'-RNA modification, both in eukaryotes and prokaryotes. The importance of 3'-oligouridylation has recently gained attention through the discovery of several types of uridylated-RNAs, by the existence of enzymes that specifically add poly(U) tails and others that preferentially degrade these tails. Namely, Dis3L2 is a 3'-5' exoribonuclease from the RNase II/RNB family that has been shown to act preferentially on oligo(U)-tailed transcripts. Our understanding of this process is still at the beginning, but it is already known to interfere in the regulation of diverse RNA species in most eukaryotes. Now that we are aware of the prevalence of RNA uridylation and the techniques available to globally evaluate the 3'-terminome, we can expect to make rapid progress in determining the extent of terminal oligouridylation in different RNA populations and unravel its impact on RNA decay mechanisms. Here, we sum up what is known about 3'-RNA modification in the different cellular compartments of eukaryotic cells, the conserved enzymes that perform this 3'-end modification and the effectors that are selectively activated by this process.


Subject(s)
RNA 3' End Processing , RNA/chemistry , RNA/metabolism , Animals , Cell Compartmentation , Exoribonucleases/chemistry , Exoribonucleases/metabolism , Humans , Metabolic Networks and Pathways , Models, Biological , Models, Molecular , Oligoribonucleotides/chemistry , Oligoribonucleotides/metabolism , Poly U/chemistry , Poly U/metabolism , Protein Conformation , RNA Stability , Uracil Nucleotides/chemistry , Uracil Nucleotides/metabolism
19.
EMBO J ; 34(13): 1801-15, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-25979828

ABSTRACT

Terminal uridylyl transferases (TUTs) function as integral regulators of microRNA (miRNA) biogenesis. Using biochemistry, single-molecule, and deep sequencing techniques, we here investigate the mechanism by which human TUT7 (also known as ZCCHC6) recognizes and uridylates precursor miRNAs (pre-miRNAs) in the absence of Lin28. We find that the overhang of a pre-miRNA is the key structural element that is recognized by TUT7 and its paralogues, TUT4 (ZCCHC11) and TUT2 (GLD2/PAPD4). For group II pre-miRNAs, which have a 1-nt 3' overhang, TUT7 restores the canonical end structure (2-nt 3' overhang) through mono-uridylation, thereby promoting miRNA biogenesis. For pre-miRNAs where the 3' end is further recessed into the stem (as in 3' trimmed pre-miRNAs), TUT7 generates an oligo-U tail that leads to degradation. In contrast to Lin28-stimulated oligo-uridylation, which is processive, a distributive mode is employed by TUT7 for both mono- and oligo-uridylation in the absence of Lin28. The overhang length dictates the frequency (but not duration) of the TUT7-RNA interaction, thus explaining how TUT7 differentiates pre-miRNA species with different overhangs. Our study reveals dual roles and mechanisms of uridylation in repair and removal of defective pre-miRNAs.


Subject(s)
MicroRNAs/metabolism , RNA Nucleotidyltransferases/physiology , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , Uridine Monophosphate/metabolism , Adenine Nucleotides/metabolism , Base Sequence , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Oligoribonucleotides/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA Stability/genetics , Uracil Nucleotides/metabolism
20.
Nucleic Acids Res ; 43(8): 4236-48, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25852104

ABSTRACT

The Nuclear Exosome Targeting (NEXT) complex is a key cofactor of the mammalian nuclear exosome in the removal of Promoter Upstream Transcripts (PROMPTs) and potentially aberrant forms of other noncoding RNAs, such as snRNAs. NEXT is composed of three subunits SKIV2L2, ZCCHC8 and RBM7. We have recently identified the NEXT complex in our screen for oligo(U) RNA-binding factors. Here, we demonstrate that NEXT displays preference for U-rich pyrimidine sequences and this RNA binding is mediated by the RNA recognition motif (RRM) of the RBM7 subunit. We solved the structure of RBM7 RRM and identified two phenylalanine residues that are critical for interaction with RNA. Furthermore, we showed that these residues are required for the NEXT interaction with snRNAs in vivo. Finally, we show that depletion of components of the NEXT complex alone or together with exosome nucleases resulted in the accumulation of mature as well as extended forms of snRNAs. Thus, our data suggest a new scenario in which the NEXT complex is involved in the surveillance of snRNAs and/or biogenesis of snRNPs.


Subject(s)
RNA, Small Nuclear/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Amino Acid Motifs , Base Sequence , HEK293 Cells , HeLa Cells , Humans , Oligoribonucleotides/metabolism , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA, Small Nuclear/chemistry , RNA-Binding Proteins/analysis , Uracil Nucleotides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL