Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 693
Filtrar
1.
Stem Cell Res Ther ; 14(1): 97, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-37076930

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Humanos , Feminino , Idoso , Adolescente , Adulto Jovem , Adulto , Pós-Menopausa , Células-Tronco Mesenquimais/metabolismo , Diferenciação Celular , Nucleotídeos de Uracila/metabolismo , Nucleotídeos de Uracila/farmacologia , Difosfato de Uridina/metabolismo , Difosfato de Uridina/farmacologia , Trifosfato de Adenosina/metabolismo , Células da Medula Óssea , Células Cultivadas
2.
J Immunol ; 207(5): 1275-1287, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34389624

RESUMO

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.


Assuntos
Dinoprostona/metabolismo , Inflamação/imunologia , Interleucina-6/metabolismo , Mucosa Respiratória/metabolismo , Trifosfato de Adenosina/farmacocinética , Alarminas/metabolismo , Canais de Cálcio Ativados pela Liberação de Cálcio/metabolismo , Células Cultivadas , Humanos , Imunomodulação , Interleucina-6/genética , Fatores de Transcrição NFATC/metabolismo , Fosfolipases A2/metabolismo , Receptores Purinérgicos P2X/metabolismo , Mucosa Respiratória/patologia , Transdução de Sinais , Nucleotídeos de Uracila/metabolismo
3.
J Med Chem ; 62(9): 4555-4570, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-30951311

RESUMO

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.


Assuntos
Alanina/análogos & derivados , Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Pró-Fármacos/farmacologia , Nucleotídeos de Uracila/farmacologia , Uridina/análogos & derivados , Alanina/síntese química , Alanina/farmacologia , Animais , Antivirais/síntese química , Linhagem Celular Tumoral , Cães , Hepacivirus/enzimologia , Hepatite C/tratamento farmacológico , Humanos , Inibidores da Síntese de Ácido Nucleico/síntese química , Inibidores da Síntese de Ácido Nucleico/farmacologia , Fosforamidas , Pró-Fármacos/síntese química , Replicon/efeitos dos fármacos , Nucleotídeos de Uracila/síntese química , Nucleotídeos de Uracila/metabolismo , Uridina/síntese química , Uridina/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores
4.
Brain Res Bull ; 151: 12-24, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30922852

RESUMO

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.


Assuntos
Receptores Purinérgicos P2Y/metabolismo , Receptores Purinérgicos P2Y/fisiologia , Nucleotídeos de Adenina/metabolismo , Animais , Humanos , Agregação Plaquetária , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Receptores Purinérgicos P2Y/ultraestrutura , Transdução de Sinais , Relação Estrutura-Atividade , Nucleotídeos de Uracila/metabolismo
5.
Pharmacol Ther ; 190: 24-80, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29660366

RESUMO

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.


Assuntos
Agonistas do Receptor Purinérgico P2Y/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Purinérgicos P2Y/efeitos dos fármacos , Animais , Desenvolvimento de Medicamentos/métodos , Humanos , Ligantes , Antagonistas do Receptor Purinérgico P2Y/química , Ensaio Radioligante , Receptores Purinérgicos P2Y/metabolismo , Solubilidade , Nucleotídeos de Uracila/metabolismo
6.
Sci Rep ; 5: 16861, 2015 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-26578061

RESUMO

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.


Assuntos
Desoxirribonucleotídeos/metabolismo , Fluoruracila/farmacologia , Redes e Vias Metabólicas/efeitos dos fármacos , Modelos Biológicos , Redes Neurais de Computação , Ribonucleotídeos/metabolismo , Nucleotídeos de Adenina/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida , Nucleotídeos de Citosina/metabolismo , Nucleotídeos de Guanina , Células Hep G2 , Humanos , Concentração Inibidora 50 , Espectrometria de Massas em Tandem , Nucleotídeos de Timina/metabolismo , Nucleotídeos de Uracila/metabolismo
7.
EMBO J ; 34(13): 1801-15, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-25979828

RESUMO

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.


Assuntos
MicroRNAs/metabolismo , RNA Nucleotidiltransferases/fisiologia , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA , Uridina Monofosfato/metabolismo , Nucleotídeos de Adenina/metabolismo , Sequência de Bases , Células HEK293 , Células HeLa , Humanos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Oligorribonucleotídeos/metabolismo , Processamento Pós-Transcricional do RNA/genética , Estabilidade de RNA/genética , Nucleotídeos de Uracila/metabolismo
8.
Nature ; 514(7521): 252-256, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25119025

RESUMO

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.


Assuntos
Exorribonucleases/química , Exorribonucleases/metabolismo , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Biocatálise , Domínio Catalítico , Cristalografia por Raios X , Complexo Multienzimático de Ribonucleases do Exossomo/química , Camundongos , MicroRNAs/química , MicroRNAs/genética , Modelos Moleculares , Oligorribonucleotídeos/química , Oligorribonucleotídeos/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Schizosaccharomyces pombe/química , Especificidade por Substrato , Nucleotídeos de Uracila/química , Nucleotídeos de Uracila/metabolismo
9.
PLoS One ; 8(7): e68575, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23922657

RESUMO

The secretion of angiogenic factors by vascular endothelial cells is one of the key mechanisms of angiogenesis. Here we report on the isolation of a new potent angiogenic factor, diuridine tetraphosphate (Up4U) from the secretome of human endothelial cells. The angiogenic effect of the endothelial secretome was partially reduced after incubation with alkaline phosphatase and abolished in the presence of suramin. In one fraction, purified to homogeneity by reversed phase and affinity chromatography, Up4U was identified by MALDI-LIFT-fragment-mass-spectrometry, enzymatic cleavage analysis and retention-time comparison. Beside a strong angiogenic effect on the yolk sac membrane and the developing rat embryo itself, Up4U increased the proliferation rate of endothelial cells and, in the presence of PDGF, of vascular smooth muscle cells. Up4U stimulated the migration rate of endothelial cells via P2Y2-receptors, increased the ability of endothelial cells to form capillary-like tubes and acts as a potent inducer of sprouting angiogenesis originating from gel-embedded EC spheroids. Endothelial cells released Up4U after stimulation with shear stress. Mean total plasma Up4U concentrations of healthy subjects (N=6) were sufficient to induce angiogenic and proliferative effects (1.34 ± 0.26 nmol L(-1)). In conclusion, Up4U is a novel strong human endothelium-derived angiogenic factor.


Assuntos
Indutores da Angiogênese/metabolismo , Endotélio Vascular/metabolismo , Adulto , Indutores da Angiogênese/química , Indutores da Angiogênese/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Membrana Corioalantoide/efeitos dos fármacos , Membrana Corioalantoide/embriologia , Embrião de Mamíferos/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Técnicas In Vitro , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Peso Molecular , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fosforilação/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ratos , Ratos Wistar , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Nucleotídeos de Uracila/química , Nucleotídeos de Uracila/metabolismo , Nucleotídeos de Uracila/farmacologia
10.
J Pharmacol Exp Ther ; 347(1): 38-46, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23908386

RESUMO

The orphan receptor GPR17 has been reported to be activated by UDP, UDP-sugars, and cysteinyl leukotrienes, and coupled to intracellular Ca(2+) mobilization and inhibition of cAMP accumulation, but other studies have reported either a different agonist profile or lack of agonist activity altogether. To determine if GPR17 is activated by uracil nucleotides and leukotrienes, the hemagglutinin-tagged receptor was expressed in five different cell lines and the signaling properties of the receptor were investigated. In C6, 1321N1, or Chinese hamster ovary (CHO) cells stably expressing GPR17, UDP, UDP-glucose, UDP-galactose, and cysteinyl leukotriene C4 (LTC4) all failed to promote inhibition of forskolin-stimulated cAMP accumulation, whereas both UDP and UDP-glucose promoted marked inhibition (>80%) of forskolin-stimulated cAMP accumulation in C6 and CHO cells expressing the P2Y14 receptor. Likewise, none of these compounds promoted accumulation of inositol phosphates in COS-7 or human embryonic kidney 293 cells transiently transfected with GPR17 alone or cotransfected with Gαq/i5, which links Gi-coupled receptors to the Gq-regulated phospholipase C (PLC) signaling pathway, or PLCε, which is activated by the Gα12/13 signaling pathway. Moreover, none of these compounds promoted internalization of GPR17 in 1321N1-GPR17 cells. Consistent with previous reports, coexpression experiments of GPR17 with cysteinyl leukotriene receptor 1 (CysLTR1) suggested that GPR17 acts as a negative regulator of CysLTR1. Taken together, these data suggest that UDP, UDP-glucose, UDP-galactose, and LTC4 are not the cognate ligands of GPR17.


Assuntos
Cisteína/metabolismo , Leucotrienos/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Nucleotídeos de Uracila/metabolismo , Animais , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Cricetulus , Cisteína/farmacologia , Células HEK293 , Humanos , Leucotrienos/farmacologia , Nucleotídeos de Uracila/farmacologia , Uridina Difosfato Glucose/metabolismo , Uridina Difosfato Glucose/farmacologia
11.
J Biol Chem ; 286(12): 10593-604, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21209081

RESUMO

The developing and mature central nervous system contains neural precursor cells expressing the proteoglycan NG2. Some of these cells continuously differentiate to myelin-forming oligodendrocytes; knowledge of the destiny of NG2(+) precursors would benefit from the characterization of new key functional players. In this respect, the G protein-coupled membrane receptor GPR17 has recently emerged as a new timer of oligodendrogliogenesis. Here, we used purified oligodendrocyte precursor cells (OPCs) to fully define the immunophenotype of the GPR17-expressing cells during OPC differentiation, unveil its native signaling pathway, and assess the functional consequences of GPR17 activation by its putative endogenous ligands, uracil nucleotides and cysteinyl leukotrienes (cysLTs). GPR17 presence was restricted to very early differentiation stages and completely segregated from that of mature myelin. Specifically, GPR17 decorated two subsets of slowly proliferating NG2(+) OPCs: (i) morphologically immature cells expressing other early proteins like Olig2 and PDGF receptor-α, and (ii) ramified preoligodendrocytes already expressing more mature factors, like O4 and O1. Thus, GPR17 is a new marker of these transition stages. In OPCs, GPR17 activation by either uracil nucleotides or cysLTs resulted in potent inhibition of intracellular cAMP formation. This effect was counteracted by GPR17 antagonists and receptor silencing with siRNAs. Finally, uracil nucleotides promoted and GPR17 inhibition, by either antagonists or siRNAs, impaired the normal program of OPC differentiation. These data have implications for the in vivo behavior of NG2(+) OPCs and point to uracil nucleotides and cysLTs as main extrinsic local regulators of these cells under physiological conditions and during myelin repair.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Regulação da Expressão Gênica/fisiologia , Oligodendroglia/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Células-Tronco/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Bainha de Mielina/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Fator de Transcrição 2 de Oligodendrócitos , Oligodendroglia/citologia , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/biossíntese , Células-Tronco/citologia , Nucleotídeos de Uracila/metabolismo , Nucleotídeos de Uracila/farmacologia
12.
Peptides ; 31(8): 1441-8, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20441784

RESUMO

Picornaviruses have a 22-24 amino acid peptide, VPg, bound covalently at the 5' end of their RNA, that is essential for replication. VPgs are uridylylated at a conserved tyrosine to form VPgpU, the primer of RNA synthesis by the viral polymerase. This first complete structure for any uridylylated VPg, of poliovirus type 1 (PV1)-VPgpU, shows that conserved amino acids in VPg stabilize the bound UMP, with the uridine atoms involved in base pairing and chain elongation projected outward. Comparing this structure to PV1-VPg and partial structures of VPg/VPgpU from other picornaviruses suggests that enteroviral polymerases require a more stable VPg structure than does the distantly related aphthovirus, foot and mouth disease virus (FMDV). The glutamine residue at the C-terminus of PV1-VPgpU lies in back of the uridine base and may stabilize its position during chain elongation and/or contribute to base specificity. Under in vivo-like conditions with the authentic cre(2C) hairpin RNA and Mg(2+), 5-methylUTP cannot compete with UTP for VPg uridylyation in an in vitro uridylyation assay, but both nucleotides are equally incorporated by PV1-polymerase with Mn(2+) and a poly-A RNA template. This indicates the 5 position is recognized under in vivo conditions. The compact VPgpU structure docks within the active site cavity of the PV-polymerase, close to the position seen for the fragment of FMDV-VPgpU with its polymerase. This structure could aid in design of novel enterovirus inhibitors, and stabilization upon uridylylation may also be pertinent for post-translational uridylylation reactions that underlie other biological processes.


Assuntos
Peptídeos/química , Peptídeos/metabolismo , Poliovirus/fisiologia , Nucleotídeos de Uracila/química , Proteínas Virais/química , Sequência de Aminoácidos , Antivirais/química , Ligação Competitiva , Desenho de Fármacos , Células HeLa , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Poliovirus/crescimento & desenvolvimento , Conformação Proteica , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/metabolismo , Alinhamento de Sequência , Especificidade por Substrato , Nucleotídeos de Uracila/metabolismo , Uridina Monofosfato/química , Uridina Monofosfato/metabolismo , Uridina Trifosfato/análogos & derivados , Uridina Trifosfato/metabolismo , Uridina Trifosfato/farmacologia , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos
13.
Cell Calcium ; 45(5): 485-98, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19386359

RESUMO

Nucleotides play an important role in brain development and may exert their action via ligand-gated cationic channels or G protein-coupled receptors. Patch-clamp measurements indicated that in contrast to AMPA, ATP did not induce membrane currents in human midbrain derived neuronal progenitor cells (hmNPCs). Various nucleotide agonists concentration-dependently increased [Ca(2+)](i) as measured by the Fura-2 method, with the rank order of potency ATP>ADP>UTP>UDP. A Ca(2+)-free external medium moderately decreased, whereas a depletion of the intracellular Ca(2+) storage sites by cyclopiazonic acid markedly depressed the [Ca(2+)](i) transients induced by either ATP or UTP. Further, the P2Y(1) receptor antagonistic PPADS and MRS 2179, as well as the nucleotide catalyzing enzyme apyrase, allmost abolished the effects of these two nucleotides. However, the P2Y(1,2,12) antagonistic suramin only slightly blocked the action of ATP, but strongly inhibited that of UTP. In agreement with this finding, UTP evoked the release of ATP from hmNPCs in a suramin-, but not PPADS-sensitive manner. Immunocytochemistry indicated the co-localization of P2Y(1,2,4)-immunoreactivities (IR) with nestin-IR at these cells. In conclusion, UTP may induce the release of ATP from hmNPCs via P2Y(2) receptor-activation and thereby causes [Ca(2+)](i) transients by stimulating a P2Y(1)-like receptor.


Assuntos
Nucleotídeos de Adenina/metabolismo , Cálcio/metabolismo , Mesencéfalo/citologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Nucleotídeos de Uracila/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurogênese/fisiologia , Neurônios/citologia , Técnicas de Patch-Clamp , Agonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/metabolismo , Células-Tronco/citologia
14.
Int J Biochem Cell Biol ; 40(12): 2901-13, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18656547

RESUMO

The enzyme deoxyuridine 5'-triphosphate nucleotidohydrolase (dUTPase) is responsible for the control of intracellular levels of dUTP thus controlling the incorporation of uracil into DNA during replication. Trypanosomes and certain eubacteria contain a dimeric dUTP-dUDPase belonging to the recently described superfamily of all-alpha NTP pyrophosphatases which bears no resemblance with typical eukaryotic trimeric dUTPases and presents unique properties regarding substrate specificity and product inhibition. While the biological trimeric enzymes have been studied in detail and the human enzyme has been proposed as a promising novel target for anticancer chemotherapeutic strategies, little is known regarding the biological function of dimeric proteins. Here, we show that in Trypanosoma brucei, the dimeric dUTPase is a nuclear enzyme and that down-regulation of activity by RNAi greatly reduces cell proliferation and increases the intracellular levels of dUTP. Defects in growth could be partially reverted by the addition of exogenous thymidine. dUTPase-depleted cells presented hypersensitivity to methotrexate, a drug that increases the intracellular pools of dUTP, and enhanced uracil-DNA glycosylase activity, the first step in base excision repair. The knockdown of activity produces numerous DNA strand breaks and defects in both S and G2/M progression. Multiple parasites with a single enlarged nucleus were visualized together with an enhanced population of anucleated cells. We conclude that dimeric dUTPases are strongly involved in the control of dUTP incorporation and that adequate levels of enzyme are indispensable for efficient cell cycle progression and DNA replication.


Assuntos
Ciclo Celular/fisiologia , Dano ao DNA , Pirofosfatases/metabolismo , Trypanosoma brucei brucei/metabolismo , Animais , Reparo do DNA/efeitos dos fármacos , Fase G2/fisiologia , Plasmídeos , Pirofosfatases/genética , Interferência de RNA , Fase S/fisiologia , Nucleotídeos de Timina/análise , Nucleotídeos de Timina/metabolismo , Transfecção , Trypanosoma brucei brucei/genética , Uracila/metabolismo , Uracila/farmacologia , Nucleotídeos de Uracila/análise , Nucleotídeos de Uracila/metabolismo , Uracila-DNA Glicosidase/genética , Uracila-DNA Glicosidase/metabolismo
15.
Structure ; 16(1): 82-92, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18184586

RESUMO

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.


Assuntos
Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Orotato Fosforribosiltransferase/química , Orotato Fosforribosiltransferase/metabolismo , Orotidina-5'-Fosfato Descarboxilase/química , Orotidina-5'-Fosfato Descarboxilase/metabolismo , Desenho de Fármacos , Humanos , Cinética , Modelos Moleculares , Orotidina-5'-Fosfato Descarboxilase/efeitos dos fármacos , Conformação Proteica , Nucleotídeos de Uracila/química , Nucleotídeos de Uracila/metabolismo
16.
Electrophoresis ; 28(23): 4435-46, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17987629

RESUMO

Telomerase activity is elevated in more than 85% of cancer cells and absent in most of the normal cells and thus represents a potential cancer biomarker. We report its measurement in colon and bladder cancer cells captured using antibody-coated magnetic beads. The cells are lysed and telomerase activity is detected using a biosensor assay that employs an oligonucleotide containing the telomerase recognition sequence also covalently coupled to magnetic beads. Telomerase activity is measured by the incorporation of multiple biotinylated nucleotides at the 3'-end of the oligonucleotide strands during elongation which are then reacted with streptavidin-conjugated horseradish peroxidase. A luminescent signal is generated when hydrogen peroxidase is added in the presence of luminol and a signal enhancer. LOD experiments confirm sensitivity down to ten cancer cell equivalents. The telomerase assay reliably identified patient samples considered by an independent pathological review to contain cancer cells. Samples from normal healthy volunteers were all telomerase negative. The assay, which is amenable to automation, demonstrated high sensitivity and specificity in a small clinical cohort, making it of potential benefit as a first line assay for detection and monitoring of colon and bladder cancer.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias do Colo/enzimologia , Separação Imunomagnética/métodos , Proteínas de Neoplasias/análise , Telomerase/análise , Neoplasias da Bexiga Urinária/enzimologia , Proteínas de Bactérias/metabolismo , Técnicas Biossensoriais/métodos , Biotina/química , Biotina/metabolismo , Neoplasias do Colo/diagnóstico , Neoplasias do Colo/patologia , Fezes/citologia , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Medições Luminescentes , Proteínas de Neoplasias/metabolismo , Sondas de Oligonucleotídeos/análise , Padrões de Referência , Sensibilidade e Especificidade , Coloração e Rotulagem , Telomerase/urina , Nucleotídeos de Uracila/química , Nucleotídeos de Uracila/metabolismo , Neoplasias da Bexiga Urinária/diagnóstico , Neoplasias da Bexiga Urinária/patologia , Urina/citologia
17.
J Physiol ; 584(Pt 2): 419-35, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17702822

RESUMO

Extracellular ATP is a potent surfactant secretagogue but its origin in the alveolus, its mechanism(s) of release and its regulatory pathways remain unknown. Previously, we showed that hypotonic swelling of alveolar A549 cells induces Ca(2+)-dependent secretion of several adenosine and uridine nucleotides, implicating regulated exocytosis. In this study, we examined sources of Ca(2+) for the elevation of intracellular Ca(2+) concentration ([Ca(2+)](i)) evoked by acute 50% hypotonic stress and the role of autocrine purinergic signalling in Ca(2+)-dependent ATP release. We found that ATP release does not directly involve Ca(2+) influx from extracellular spaces, but depends entirely on Ca(2+) mobilization from intracellular stores. The [Ca(2+)](i) response consisted of slowly rising elevation, representing mobilization from thapsigargin (TG)-insensitive stores and a superimposed rapid spike due to Ca(2+) release from TG-sensitive endoplasmic reticulum (ER) Ca(2+) stores. The latter could be abolished by hydrolysis of extracellular triphospho- and diphosphonucleotides with apyrase; blocking P2Y(2)/P2Y(6) receptors of A549 cells with suramin; blocking UDP receptors (P2Y(6)) with pyridoxal phosphate 6-azophenyl-2',4'-disulfonic acid (PPADS); emptying TG-sensitive stores downstream with TG or caffeine in Ca(2+)-free extracellular solution; or blocking the Ca(2+)-release inositol 1,4,5-triphosphate receptor channel of the ER with 2-aminoethyldiphenylborinate. These data demonstrate that the rapid [Ca(2+)](i) spike results from the autocrine stimulation of IP(3)/Ca(2+)-coupled P2Y, predominantly P2Y(6), receptors, accounting for approximately 70% of total Ca(2+)-dependent ATP release evoked by hypotonic shock. Our study reveals a novel paradigm in which stress-induced ATP release from alveolar cells is amplified by the synergistic autocrine/paracrine action of coreleased uridine and adenosine nucleotides. We suggest that a similar mechanism of purinergic signal propagation operates in other cell types.


Assuntos
Trifosfato de Adenosina/metabolismo , Comunicação Autócrina , Sinalização do Cálcio , Cálcio/metabolismo , Células Epiteliais/metabolismo , Neoplasias Pulmonares/metabolismo , Comunicação Parácrina , Nucleotídeos de Uracila/metabolismo , Apirase/farmacologia , Cafeína/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Linhagem Celular Tumoral , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Humanos , Hidrólise , Soluções Hipotônicas , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Neoplasias Pulmonares/patologia , Pressão Osmótica , Antagonistas do Receptor Purinérgico P2 , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacologia , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y2 , Suramina/farmacologia , Tapsigargina/farmacologia , Fatores de Tempo
18.
Science ; 314(5807): 1893, 2006 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-17185594

RESUMO

Double-stranded RNA, processed to small interfering RNAs (siRNAs) by Dicer and incorporated into the RNA-induced silencing complex (RISC), triggers gene silencing by a variety of pathways in eukaryotes. RNA interference involving the degradation of homologous transcripts is the best-characterized mechanism. However, the fate of the RNA fragments resulting from siRNA-directed cleavage is poorly understood. We have identified a gene (MUT68) in the unicellular green alga Chlamydomonas reinhardtii that is required for the efficient decay of siRNA-targeted transcripts. MUT68 encodes a noncanonical polyadenylate polymerase that adds untemplated adenines to the 5' RNA fragments after siRNA-mediated cleavage and appears to stimulate their exosome-dependent degradation.


Assuntos
Nucleotídeos de Adenina/metabolismo , Chlamydomonas reinhardtii/genética , Chlamydomonas reinhardtii/metabolismo , Oligorribonucleotídeos/metabolismo , Polinucleotídeo Adenililtransferase/metabolismo , RNA Mensageiro/metabolismo , Complexo de Inativação Induzido por RNA/metabolismo , Animais , Chlamydomonas reinhardtii/enzimologia , Exorribonucleases/metabolismo , Dados de Sequência Molecular , Polinucleotídeo Adenililtransferase/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Moldes Genéticos , Transgenes , Triptofano Sintase/genética , Nucleotídeos de Uracila/metabolismo
19.
EMBO J ; 25(19): 4615-27, 2006 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-16990797

RESUMO

Nucleotides and cysteinyl-leukotrienes (CysLTs) are unrelated signaling molecules inducing multiple effects through separate G-protein-coupled receptors: the P2Y and the CysLT receptors. Here we show that GPR17, a Gi-coupled orphan receptor at intermediate phylogenetic position between P2Y and CysLT receptors, is specifically activated by both families of endogenous ligands, leading to both adenylyl cyclase inhibition and intracellular calcium increases. Agonist-response profile, as determined by [(35)S]GTPgammaS binding, was different from that of already known CysLT and P2Y receptors, with EC(50) values in the nanomolar and micromolar range, for CysLTs and uracil nucleotides, respectively. Both rat and human receptors are highly expressed in the organs typically undergoing ischemic damage, that is, brain, heart and kidney. In vivo inhibition of GPR17 by either CysLT/P2Y receptor antagonists or antisense technology dramatically reduced ischemic damage in a rat focal ischemia model, suggesting GPR17 as the common molecular target mediating brain damage by nucleotides and CysLTs. In conclusion, the deorphanization of GPR17 revealed a dualistic receptor for two endogenous unrelated ligand families. These findings may lead to dualistic drugs of previously unexplored therapeutic potential.


Assuntos
Cisteína/metabolismo , Leucotrienos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Nucleotídeos de Uracila/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/induzido quimicamente , Células COS , Chlorocebus aethiops , Perfilação da Expressão Gênica , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/química , Receptores de Leucotrienos/metabolismo , Receptores Purinérgicos P2/metabolismo , Análise de Sequência de Proteína
20.
Curr Med Chem ; 13(3): 289-312, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16475938

RESUMO

Pyrimidine nucleotides, including UTP, UDP and UDP-glucose, are important signaling molecules which activate G protein-coupled membrane receptors (GPCRs) of the P2Y family. Four distinct pyrimidine nucleotide-sensitive P2Y receptor subtypes have been cloned, P2Y2, P2Y4, P2Y6 and P2Y14. P2Y2 and P2Y4 receptors are activated by UTP (the P2Y2, and the rat but not the human P2Y4 receptor are also activated by ATP), the P2Y6 receptor is activated by UDP, and the P2Y14 receptor by UDP-glucose. Furthermore, non-P2Y GPCRs, the cysteinylleukotriene receptors (CysLT1R and CysLT2R) have been described to be activated by UDP in addition to activation by cysteinylleukotrienes. While P2Y2, P2Y4, and P2Y6 receptor activation results in stimulation of phospholipase C, the P2Y14 receptor is coupled to inhibition of adenylate cyclase. Derivatives and analogs of the physiological nucleotides UTP, UDP and ATP have been synthesized and evaluated in order to obtain enzymatically stable, subtype-selective agonists. The P2Y2 receptor agonists diuridine tetraphosphate (diquafosol) and the uracil-cytosine dinucleotide denufosol are currently undergoing clinical trials for dry eye disease, retinal detachment disease, upper respiratory tract symptoms, and cystic fibrosis, respectively. The first antagonists for P2Y2 and P2Y6 receptors that appear to be selective versus other P2Y receptor subtypes have recently been described. Selective antagonists for P2Y4 and P2Y14 receptors are still lacking. Uracil nucleotide-sensitive P2Y receptor subtypes may constitute future targets for the treatment of certain cancer types, vascular diseases, inflammatory diseases, and immunomodulatory intervention. They have also been proposed to play a role in neurodegenerative diseases. This article is an updated version of "P2-Pyrimidinergic Receptors and Their Ligands" by C. E. Müller published in Curr. Pharm. Des. 2002, 8, 2353-2369.


Assuntos
Agonistas do Receptor Purinérgico P2 , Nucleotídeos de Uracila/farmacologia , Osso e Ossos/metabolismo , Fibrose Cística/tratamento farmacológico , Síndromes do Olho Seco/tratamento farmacológico , Humanos , Hipersensibilidade/tratamento farmacológico , Inflamação/tratamento farmacológico , Pneumopatias Obstrutivas/tratamento farmacológico , Neoplasias/tratamento farmacológico , Conformação de Ácido Nucleico , Antagonistas do Receptor Purinérgico P2 , Nucleotídeos de Uracila/química , Nucleotídeos de Uracila/isolamento & purificação , Nucleotídeos de Uracila/metabolismo , Nucleotídeos de Uracila/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA