Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 335
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomolecules ; 14(6)2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38927084

RESUMEN

Clickable nucleosides, most often 5-ethynyl-2'-deoxyuridine (EtU), are widely used in studies of DNA replication in living cells and in DNA functionalization for bionanotechology applications. Although clickable dNTPs are easily incorporated by DNA polymerases into the growing chain, afterwards they might become targets for DNA repair systems or interfere with faithful nucleotide insertion. Little is known about the possibility and mechanisms of these post-synthetic events. Here, we investigated the repair and (mis)coding properties of EtU and two bulkier clickable pyrimidine nucleosides, 5-(octa-1,7-diyn-1-yl)-U (C8-AlkU) and 5-(octa-1,7-diyn-1-yl)-C (C8-AlkC). In vitro, EtU and C8-AlkU, but not C8-AlkC, were excised by SMUG1 and MBD4, two DNA glycosylases from the base excision repair pathway. However, when placed into a plasmid encoding a fluorescent reporter inactivated by repair in human cells, EtU and C8-AlkU persisted for much longer than uracil or its poorly repairable phosphorothioate-flanked derivative. DNA polymerases from four different structural families preferentially bypassed EtU, C8-AlkU and C8-AlkC in an error-free manner, but a certain degree of misincorporation was also observed, especially evident for DNA polymerase ß. Overall, clickable pyrimidine nucleotides could undergo repair and be a source of mutations, but the frequency of such events in the cell is unlikely to be considerable.


Asunto(s)
Química Clic , Reparación del ADN , Nucleótidos de Pirimidina , Humanos , Nucleótidos de Pirimidina/química , Nucleótidos de Pirimidina/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Desoxiuridina/análogos & derivados , Desoxiuridina/química , Desoxiuridina/metabolismo , ADN/metabolismo , ADN/química , ADN/genética , Replicación del ADN , Uracil-ADN Glicosidasa/metabolismo
2.
Chemistry ; 30(24): e202400137, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38403849

RESUMEN

Besides being a key player in numerous fundamental biological processes, RNA also represents a versatile platform for the creation of therapeutic agents and efficient vaccines. The production of RNA oligonucleotides, especially those decorated with chemical modifications, cannot meet the exponential demand. Due to the inherent limits of solid-phase synthesis and in vitro transcription, alternative, biocatalytic approaches are in dire need to facilitate the production of RNA oligonucleotides. Here, we present a first step towards the controlled enzymatic synthesis of RNA oligonucleotides. We have explored the possibility of a simple protection step of the vicinal cis-diol moiety to temporarily block ribonucleotides. We demonstrate that pyrimidine nucleotides protected with acetals, particularly 2',3'-O-isopropylidene, are well-tolerated by the template-independent RNA polymerase PUP (polyU polymerase) and highly efficient coupling reactions can be achieved within minutes - an important feature for the development of enzymatic de novo synthesis protocols. Even though purines are not equally well-tolerated, these findings clearly demonstrate the possibility of using cis-diol-protected ribonucleotides combined with template-independent polymerases for the stepwise construction of RNA oligonucleotides.


Asunto(s)
ARN Polimerasas Dirigidas por ADN , ARN , ARN/química , ARN/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/química , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Oligonucleótidos/síntesis química , Ribonucleótidos/química , Ribonucleótidos/metabolismo , Nucleótidos/química , Nucleótidos/metabolismo , Nucleótidos de Pirimidina/química , Nucleótidos de Pirimidina/metabolismo
4.
Cells ; 11(3)2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35159133

RESUMEN

The etiology of dry mouth conditions is multi-faceted. Patients radiated after head and neck cancer (HNC) and those with primary Sjögren's syndrome (pSS) share many of the same symptoms despite different causes. With the aim of better understanding the pathophysiology and biochemical processes behind dry mouth with different etiologies, we investigated the metabolic profile of 10 HNC patients, 9 pSS patients and 10 healthy controls using high-performance liquid chromatography-high resolution mass spectrometry (HPLC-MS) metabolomics. Principal component analysis (PCA) revealed different metabolic profiles when comparing all subjects included in the study. Both patient groups showed higher ratios of several pyrimidine nucleotides and nucleosides when compared to controls. This finding may indicate that purinergic signaling plays a role in dry mouth conditions. Moreover, significantly increased levels of DL-3-aminoisobutyric acid were found in HNC patients when compared to controls, and a similar tendency was observed in the pSS patients. Furthermore, a dysregulation in amino acid metabolism was observed in both patient groups. In conclusion, metabolomics analysis showed separate metabolic profiles for HNC and pSS patients as compared to controls that could be useful in diagnostics and for elucidating the different pathophysiologies. The demonstrated dysregulation of pyrimidine nucleotides and levels of metabolites derived from amino acids in the patient groups should be studied further.


Asunto(s)
Neoplasias de Cabeza y Cuello , Síndrome de Sjögren , Xerostomía , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Metabolómica , Nucleótidos de Pirimidina/análisis , Nucleótidos de Pirimidina/metabolismo , Saliva/metabolismo , Síndrome de Sjögren/metabolismo , Xerostomía/metabolismo
5.
Purinergic Signal ; 17(4): 693-704, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34403084

RESUMEN

Human ecto-5-nucleotidase (CD73) is involved in purinergic signalling, which influences a diverse range of biological processes. CD73 hydrolyses AMP and is the major control point for the levels of extracellular adenosine. Inhibitors of CD73 thus block the immunosuppressive action of adenosine, a promising approach for cancer immunotherapy. Interestingly, ADP and ATP are competitive inhibitors of CD73, with the most potent small-molecule inhibitors to date being non-hydrolysable ADP analogues. While AMP is the major substrate of the enzyme, CD73 has been reported to hydrolyse other 5'-nucleoside monophosphates. Based on a fragment screening campaign at the BESSY II synchrotron, we present the binding modes of various deoxyribo- and ribonucleoside monophosphates and of four additional fragments binding to the nucleoside binding site of the open form of the enzyme. Kinetic analysis of monophosphate hydrolysis shows that ribonucleotide substrates are favoured over their deoxyribose equivalents with AMP being the best substrate. We characterised the initial step of AMP hydrolysis, the binding mode of AMP to the open conformation of CD73 and compared that to other monophosphate substrates. In addition, the inhibitory activity of various bisphosphonic acid derivatives of nucleoside diphosphates was determined. Although AMPCP remains the most potent inhibitor, replacement of the adenine base with other purines or with pyrimidines increases the Ki value only between twofold and sixfold. On the other hand, these nucleobases offer new opportunities to attach substituents for improved pharmacological properties.


Asunto(s)
5'-Nucleotidasa/metabolismo , Adenosina/metabolismo , Purinas/metabolismo , Nucleótidos de Pirimidina/metabolismo , Transducción de Señal/fisiología , Humanos , Hidrólisis , Unión Proteica , Pliegue de Proteína
7.
J Biol Chem ; 297(3): 101018, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34331944

RESUMEN

The coronaviral nonstructural protein 9 (Nsp9) is essential for viral replication; it is the primary substrate of Nsp12's pseudokinase domain within the viral replication transcription complex, an association that also recruits other components during different stages of RNA reproduction. In the unmodified state, Nsp9 forms an obligate homodimer via an essential GxxxG protein-interaction motif, but its ssRNA-binding mechanism remains unknown. Using structural biological techniques, here we show that a base-mimicking compound identified from a small molecule fragment screen engages Nsp9 via a tetrameric Pi-Pi stacking interaction that induces the formation of a parallel trimer-of-dimers. This oligomerization mechanism allows an interchange of "latching" N-termini, the charges of which contribute to a series of electropositive channels that suggests a potential interface for viral RNA. The identified pyrrolo-pyrimidine compound may also serve as a potential starting point for the development of compounds seeking to probe Nsp9's role within SARS-CoV-2 replication.


Asunto(s)
COVID-19/virología , Nucleótidos de Pirimidina/metabolismo , Proteínas de Unión al ARN/metabolismo , SARS-CoV-2/metabolismo , Proteínas Virales/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Unión Proteica , ARN/metabolismo , SARS-CoV-2/fisiología , Replicación Viral
8.
Biochem Biophys Res Commun ; 557: 117-121, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33862454

RESUMEN

Caenorhabditis elegans T09F3.2 is a homolog of the human mitochondrial pyrimidine nucleotide transporter. We isolated a T09F3.2 mutant (TOG2) with a 0.7 kb deletion in T09F3.2, which exhibited low growth and movement. TOG2 worms exhibited high glucose content and low lipid content in intestinal cells and oocytes, suggesting glucose leakage from these cells. The glucose transport inhibitor phloretin improved the growth of TOG2 worms, suggesting that T09F3.2 regulates the phloretin-sensitive glucose transporter FGT-1. The localization of T09F3.2 was examined to assess the regulation of FGT-1 by T09F3.2. Distinct expression of T09F3.2 fused with DsRed-Monomer (T09F3.2:DsRed-Monomer) was observed in the basal domain of intestinal cells and was weakly expressed in many cells. Colocalization of FGT-1 and T09F3.2 was observed in the intestinal cell surface and body wall muscle. This colocalization supports the regulation of FGT-1 by T09F3.2. These results reveal new aspects of glucose transporter regulation.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Glucosa/metabolismo , Mitocondrias/metabolismo , Nucleótidos de Pirimidina/metabolismo , Animales , Transporte Biológico , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Humanos , Intestinos/fisiología , Oocitos/metabolismo
9.
Nat Metab ; 3(5): 636-650, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33903774

RESUMEN

Cytosolic mitochondrial DNA (mtDNA) elicits a type I interferon response, but signals triggering the release of mtDNA from mitochondria remain enigmatic. Here, we show that mtDNA-dependent immune signalling via the cyclic GMP-AMP synthase‒stimulator of interferon genes‒TANK-binding kinase 1 (cGAS-STING-TBK1) pathway is under metabolic control and is induced by cellular pyrimidine deficiency. The mitochondrial protease YME1L preserves pyrimidine pools by supporting de novo nucleotide synthesis and by proteolysis of the pyrimidine nucleotide carrier SLC25A33. Deficiency of YME1L causes inflammation in mouse retinas and in cultured cells. It drives the release of mtDNA and a cGAS-STING-TBK1-dependent inflammatory response, which requires SLC25A33 and is suppressed upon replenishment of cellular pyrimidine pools. Overexpression of SLC25A33 is sufficient to induce immune signalling by mtDNA. Similarly, depletion of cytosolic nucleotides upon inhibition of de novo pyrimidine synthesis triggers mtDNA-dependent immune responses in wild-type cells. Our results thus identify mtDNA release and innate immune signalling as a metabolic response to cellular pyrimidine deficiencies.


Asunto(s)
ADN Mitocondrial/genética , Inmunidad Innata , Mitocondrias/genética , Mitocondrias/metabolismo , Nucleótidos de Pirimidina/metabolismo , Animales , Citosol/metabolismo , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Ratones , Modelos Biológicos , Nucleotidiltransferasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
10.
Appl Microbiol Biotechnol ; 104(15): 6659-6667, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32500270

RESUMEN

Polyphosphosphate kinases (PPKs) catalyse the reversible transfer of the γ-phosphate group of a nucleoside-triphosphate to a growing chain of polyphosphate. Most known PPKs are specific for ATP, but some can also use GTP as a phosphate donor. In this study, we describe the properties of a PPK2-type PPK of the ß-proteobacterium Ralstonia eutropha. The purified enzyme (PPK2c) is highly unspecific and accepts purine nucleotides as well as the pyridine nucleotides including UTP as substrates. The presence of a polyP primer is not necessary for activity. The corresponding nucleoside diphosphates and microscopically detectable polyphosphate granules were identified as reaction products. PPK2c also catalyses the formation of ATP, GTP, CTP, dTTP and UTP from the corresponding nucleoside diphosphates, if polyP is present as a phosphate donor. Remarkably, the nucleoside-tetraphosphates AT(4)P, GT(4)P, CT(4)P, dTT(4)P and UT(4)P were also detected in substantial amounts. The low nucleotide specificity of PPK2c predestines this enzyme in combination with polyP to become a powerful tool for the regeneration of ATP and other nucleotides in biotechnological applications. As an example, PPK2c and polyP were used to replace ATP and to fuel the hexokinase-catalysed phosphorylation of glucose with only catalytic amounts of ADP. KEY POINTS: • PPK2c of R. eutropha can be used for regeneration of any NTP or dNTP. • PPK2c is highly unspecific and accepts all purine and pyrimidine nucleotides. • PPK2c forms polyphosphate granules in vitro from any NTP.


Asunto(s)
Cupriavidus necator/enzimología , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Nucleótidos de Purina/metabolismo , Nucleótidos de Pirimidina/metabolismo , Uridina Difosfato/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Cupriavidus necator/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Fosforilación , Fosfotransferasas (Aceptor del Grupo Fosfato)/genética
11.
Arch Microbiol ; 202(6): 1551-1557, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32125450

RESUMEN

The control of pyrimidine nucleotide formation in the bacterium Pseudomonas aurantiaca ATCC 33663 by pyrimidines was studied. The activities of the pyrimidine biosynthetic pathway enzymes were investigated in P. aurantiaca ATCC 33663 cells and from cells of an auxotroph lacking orotate phosphoribosyltransferase activity under selected culture conditions. All activities of the pyrimidine biosynthetic pathway enzymes in ATCC 33663 cells were depressed by uracil addition to the minimal medium when succinate served as the carbon source. In contrast, all pyrimidine biosynthetic pathway enzyme activities in ATCC 33663 cells were depressed by orotic acid supplementation to the minimal medium when glucose served as the carbon source. The orotidine 5'-monophosphate decarboxylase activity in the phosphoribosyltransferase mutant strain increased by more than sixfold in succinate-grown cells and by more than 16-fold in glucose-grown cells after pyrimidine limitation showing possible repression of the decarboxylase by a pyrimidine-related compound. Inhibition by ATP, GTP, UTP and pyrophosphate of the in vitro activity of aspartate transcarbamoylase in ATCC 33663 was observed. The findings demonstrated control at the level of pyrimidine biosynthetic enzyme synthesis and activity for the P. aurantiaca transcarbamoylase. The control of pyrimidine synthesis in P. aurantiaca seemed to differ from what has been observed previously for the regulation of pyrimidine biosynthesis in related Pseudomonas species. This investigation could prove helpful to future work studying pseudomonad taxonomic analysis as well as to those exploring antifungal and antimicrobial agents produced by P. aurantiaca.


Asunto(s)
Aspartato Carbamoiltransferasa/metabolismo , Pseudomonas/metabolismo , Nucleótidos de Pirimidina/biosíntesis , Pirimidinas/metabolismo , Aspartato Carbamoiltransferasa/genética , Vías Biosintéticas , Difosfatos , Regulación Bacteriana de la Expresión Génica , Orotato Fosforribosiltransferasa/genética , Orotidina-5'-Fosfato Descarboxilasa/metabolismo , Pseudomonas/enzimología , Nucleótidos de Pirimidina/metabolismo , Ácido Succínico/metabolismo , Uracilo/metabolismo
12.
Am J Physiol Endocrinol Metab ; 316(5): E852-E865, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30860875

RESUMEN

Cancer-associated cachexia reduces survival, which has been attenuated by blocking the activin receptor type 2B (ACVR2B) ligands in mice. The purpose of this study was to unravel the underlying physiology and novel cachexia biomarkers by use of the colon-26 (C26) carcinoma model of cancer cachexia. Male BALB/c mice were subcutaneously inoculated with C26 cancer cells or vehicle control. Tumor-bearing mice were treated with vehicle (C26+PBS) or soluble ACVR2B either before (C26+sACVR/b) or before and after (C26+sACVR/c) tumor formation. Skeletal muscle and serum metabolomics analysis was conducted by gas chromatography-mass spectrometry. Cancer altered various biologically functional groups representing 1) amino acids, 2) energy sources, and 3) nucleotide-related intermediates. Muscle metabolomics revealed increased content of free phenylalanine in cancer that strongly correlated with the loss of body mass within the last 2 days of the experiment. This correlation was also detected in serum. Decreased ribosomal RNA content and phosphorylation of a marker of pyrimidine synthesis revealed changes in nucleotide metabolism in cancer. Overall, the effect of the experimental C26 cancer predominated over blocking ACVR2B ligands in both muscle and serum. However, the level of methyl phosphate, which was decreased in muscle in cancer, was restored by sACVR2B-Fc treatment. In conclusion, experimental cancer affected muscle and blood metabolomes mostly independently of blocking ACVR2B ligands. Of the affected metabolites, we have identified free phenylalanine as a promising biomarker of muscle atrophy or cachexia. Finally, the decreased capacity for pyrimidine nucleotide and protein synthesis in tumor-bearing mice opens up new avenues in cachexia research.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Caquexia/metabolismo , Neoplasias del Colon/metabolismo , Metaboloma/fisiología , Músculo Esquelético/metabolismo , Aminoácidos/metabolismo , Animales , Caquexia/etiología , Línea Celular Tumoral , Neoplasias del Colon/complicaciones , Fragmentos Fc de Inmunoglobulinas/farmacología , Masculino , Redes y Vías Metabólicas , Metaboloma/efectos de los fármacos , Ratones , Músculo Esquelético/efectos de los fármacos , Organofosfatos/metabolismo , Fenilalanina/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/fisiología , Nucleótidos de Pirimidina/metabolismo , Proteínas Recombinantes
13.
Trends Mol Med ; 24(10): 886-903, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30120023

RESUMEN

Innovations in epitranscriptomics have resulted in the identification of more than 160 RNA modifications to date. These developments, together with the recent discovery of writers, readers, and erasers of modifications occurring across a wide range of RNAs and tissue types, have led to a surge in integrative approaches for transcriptome-wide mapping of modifications and protein-RNA interaction profiles of epitranscriptome players. RNA modification maps and crosstalk between them have begun to elucidate the role of modifications as signaling switches, entertaining the notion of an epitranscriptomic code as a driver of the post-transcriptional fate of RNA. Emerging single-molecule sequencing technologies and development of antibodies specific to various RNA modifications could enable charting of transcript-specific epitranscriptomic marks across cell types and their alterations in disease.


Asunto(s)
Epigénesis Genética , Nucleótidos de Purina/metabolismo , Nucleótidos de Pirimidina/metabolismo , Procesamiento Postranscripcional del ARN , ARN/genética , Transcriptoma , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Anomalías Congénitas/genética , Anomalías Congénitas/metabolismo , Anomalías Congénitas/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Metilación , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Nucleótidos de Purina/genética , Nucleótidos de Pirimidina/genética , ARN/clasificación , ARN/metabolismo
14.
Microcirculation ; 25(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29110409

RESUMEN

Smooth muscle voltage-gated potassium (Kv) channels are important regulators of microvascular tone and tissue perfusion. Recent studies indicate that Kv1 channels represent a key component of the physiological coupling between coronary blood flow and myocardial oxygen demand. While the mechanisms by which metabolic changes in the heart are transduced to alter coronary Kv1 channel gating and promote vasodilation are unclear, a growing body of evidence underscores a pivotal role of Kv1 channels in sensing the cellular redox status. Here, we discuss current knowledge of mechanisms of Kv channel redox regulation with respect to pyridine nucleotide modulation of Kv1 function via ancillary Kvß proteins as well as direct modulation of channel activity via reactive oxygen and nitrogen species. We identify areas of additional research to address the integration of regulatory processes under altered physiological and pathophysiological conditions that may reveal insights into novel treatment strategies for conditions in which the matching of coronary blood supply and myocardial oxygen demand is compromised.


Asunto(s)
Canales de Potasio con Entrada de Voltaje/fisiología , Nucleótidos de Pirimidina/metabolismo , Animales , Vasos Coronarios/química , Humanos , Microcirculación , Oxidación-Reducción , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo
15.
Nucleic Acids Res ; 45(3): 1539-1552, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28180308

RESUMEN

The DEAH box helicase Prp43 is a bifunctional enzyme from the DEAH/RHA helicase family required both for the maturation of ribosomes and for lariat intron release during splicing. It interacts with G-patch domain containing proteins which activate the enzymatic activity of Prp43 in vitro by an unknown mechanism. In this work, we show that the activation by G-patch domains is linked to the unique nucleotide binding mode of this helicase family. The base of the ATP molecule is stacked between two residues, R159 of the RecA1 domain (R-motif) and F357 of the RecA2 domain (F-motif). Using Prp43 F357A mutants or pyrimidine nucleotides, we show that the lack of stacking of the nucleotide base to the F-motif decouples the NTPase and helicase activities of Prp43. In contrast the R159A mutant (R-motif) showed reduced ATPase and helicase activities. We show that the Prp43 R-motif mutant induces the same phenotype as the absence of the G-patch protein Gno1, strongly suggesting that the processing defects observed in the absence of Gno1 result from a failure to activate the Prp43 helicase. Overall we propose that the stacking between the R- and F-motifs and the nucleotide base is important for the activity and regulation of this helicase family.


Asunto(s)
Adenosina Trifosfato/metabolismo , ARN Helicasas DEAD-box/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Adenosina Trifosfato/química , Sustitución de Aminoácidos , Dominio Catalítico/genética , Cristalografía por Rayos X , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , Activación Enzimática , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Dominios y Motivos de Interacción de Proteínas , Nucleótidos de Pirimidina/química , Nucleótidos de Pirimidina/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
16.
J Biomol Struct Dyn ; 35(10): 2136-2154, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27376462

RESUMEN

Thymidylate kinase (TMK) is a key enzyme for the synthesis of DNA, making it an important target for the development of anticancer, antibacterial, and antiparasitic drugs. TMK homologs exhibit significant variations in sequence, residue conformation, substrate specificity, and oligomerization mode. However, the influence of sequence evolution and conformational dynamics on its quaternary structure and function has not been studied before. Based on extensive sequence and structure analyses, our study detected several non-conserved residues which are linked by co-evolution and are implicated in the observed variations in flexibility, oligomeric assembly, and substrate specificity among the homologs. These lead to differences in the pattern of interactions at the active site in TMKs of different specificity. The method was further tested on TMK from Sulfolobus tokodaii (StTMK) which has substantial differences in sequence and structure compared to other TMKs. Our analyses pointed to a more flexible dTMP-binding site in StTMK compared to the other homologs. Binding assays proved that the protein can accommodate both purine and pyrimidine nucleotides at the dTMP binding site with comparable affinity. Additionally, the residues responsible for the narrow specificity of Brugia malayi TMK, whose three-dimensional structure is unavailable, were detected. Our study provides a residue-level understanding of the differences observed among TMK homologs in previous experiments. It also illustrates the correlation among sequence evolution, conformational dynamics, oligomerization mode, and substrate recognition in TMKs and detects co-evolving residues that affect binding, which should be taken into account while designing novel inhibitors.


Asunto(s)
Proteínas Arqueales/química , Brugia Malayi/química , Proteínas del Helminto/química , Nucleósido-Fosfato Quinasa/química , Nucleótidos de Purina/química , Nucleótidos de Pirimidina/química , Sulfolobus/química , Secuencia de Aminoácidos , Animales , Proteínas Arqueales/metabolismo , Sitios de Unión , Brugia Malayi/enzimología , Cristalografía por Rayos X , Proteínas del Helminto/metabolismo , Humanos , Cinética , Simulación de Dinámica Molecular , Nucleósido-Fosfato Quinasa/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Nucleótidos de Purina/metabolismo , Nucleótidos de Pirimidina/metabolismo , Alineación de Secuencia , Homología Estructural de Proteína , Especificidad por Sustrato , Sulfolobus/enzimología , Termodinámica
17.
Biochimie ; 121: 161-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26700143

RESUMEN

Reactive oxygen species damage DNA bases to produce 7,8-dihydro-8-oxo-2'-deoxyguanosine (8-oxoG), which results in G:C to T:A transversions. To better understand mechanisms of dNTP incorporation opposite 8-oxoG, we performed pre-steady-state kinetic analysis of nucleotide incorporation using the catalytic core of yeast DNA polymerase η (Pol ηcore, residues 1-513) instead of full-length Pol η, eliminating potential effects of the C-terminal C2H2 sequence motif on dNTP incorporation. Kinetic analysis showed that Pol ηcore preferred to incorporate dCTP opposite 8-oxoG. A lack of a pre-steady-state kinetic burst for Pol ηcore suggested that dCTP incorporation is slower than the dissociation of the polymerase from DNA. The extension products beyond the 8-oxoG were determined by LC-MS/MS and showed that 57% of the products corresponded to the correct incorporation (C) and 43% corresponded to dATP misincorporation. More dATP was incorporated opposite 8-oxoG with a mixture of dNTPs than predicted using only a single dNTP. The kinetic analysis of 8-oxoG bypass by yeast DNA Pol ηcore provides further understanding of the mechanism of mutation at this oxidation lesion with yeast DNA polymerase η.


Asunto(s)
ADN Polimerasa Dirigida por ADN/metabolismo , Desoxiguanosina/análogos & derivados , 8-Hidroxi-2'-Desoxicoguanosina , Dominio Catalítico , Desoxiguanosina/química , Desoxiguanosina/metabolismo , Cinética , Nucleótidos de Pirimidina/metabolismo
18.
J Biol Chem ; 289(48): 33137-48, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25320081

RESUMEN

The human genome encodes 53 members of the solute carrier family 25 (SLC25), also called the mitochondrial carrier family, many of which have been shown to transport inorganic anions, amino acids, carboxylates, nucleotides, and coenzymes across the inner mitochondrial membrane, thereby connecting cytosolic and matrix functions. Here two members of this family, SLC25A33 and SLC25A36, have been thoroughly characterized biochemically. These proteins were overexpressed in bacteria and reconstituted in phospholipid vesicles. Their transport properties and kinetic parameters demonstrate that SLC25A33 transports uracil, thymine, and cytosine (deoxy)nucleoside di- and triphosphates by an antiport mechanism and SLC25A36 cytosine and uracil (deoxy)nucleoside mono-, di-, and triphosphates by uniport and antiport. Both carriers also transported guanine but not adenine (deoxy)nucleotides. Transport catalyzed by both carriers was saturable and inhibited by mercurial compounds and other inhibitors of mitochondrial carriers to various degrees. In confirmation of their identity (i) SLC25A33 and SLC25A36 were found to be targeted to mitochondria and (ii) the phenotypes of Saccharomyces cerevisiae cells lacking RIM2, the gene encoding the well characterized yeast mitochondrial pyrimidine nucleotide carrier, were overcome by expressing SLC25A33 or SLC25A36 in these cells. The main physiological role of SLC25A33 and SLC25A36 is to import/export pyrimidine nucleotides into and from mitochondria, i.e. to accomplish transport steps essential for mitochondrial DNA and RNA synthesis and breakdown.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Nucleótidos de Pirimidina/química , Nucleótidos de Pirimidina/metabolismo , Animales , Transporte Biológico Activo/fisiología , Células CHO , Cricetinae , Cricetulus , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Humanos , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Nucleótidos/química , Proteínas de Transporte de Nucleótidos/genética , Proteínas de Transporte de Nucleótidos/metabolismo , ARN/genética , ARN/metabolismo , ARN Mitocondrial , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
20.
Purinergic Signal ; 10(2): 241-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24310605

RESUMEN

Receptors for purines and pyrimidines are expressed throughout the cardiovascular system. This study investigated their functional expression in porcine isolated pancreatic arteries. Pancreatic arteries (endothelium intact or denuded) were prepared for isometric tension recording and preconstricted with U46619, a thromboxane A(2) mimetic; adenosine-5'-diphosphate (ADP), uridine-5'-triphosphate (UTP) and MRS2768, a selective P2Y(2) agonist, were applied cumulatively, while adenosine-5'-triphosphate (ATP) and αß-methylene-ATP (αß-meATP) response curves were generated from single concentrations per tissue segment. Antagonists/enzyme inhibitors were applied prior to U46619 addition. ATP, αß-meATP, UTP and MRS2768 induced vasoconstriction, with a potency order of αß-meATP > MRS2768 > ATP ≥ UTP. Contractions to ATP and αß-meATP were blocked by NF449, a selective P2X(1) receptor antagonist. The contraction induced by ATP, but not UTP, was followed by vasorelaxation. Endothelium removal and DUP 697, a cyclooxygenase-2 inhibitor, had no significant effect on contraction to ATP but attenuated that to UTP, indicating actions at distinct receptors. MRS2578, a selective P2Y(6) receptor antagonist, had no effect on contractions to UTP. ADP induced endothelium-dependent vasorelaxation which was inhibited by MRS2179, a selective P2Y(1) receptor antagonist, or SCH58261, a selective adenosine A(2A) receptor antagonist. The contractions to ATP and αß-meATP were attributed to actions at P2X(1) receptors on the vascular smooth muscle, whereas it was shown for the first time that UTP induced an endothelium-dependent vasoconstriction which may involve P2Y(2) and/or P2Y(4) receptors. The relaxation induced by ADP is mediated by P2Y(1) and A(2A) adenosine receptors. Porcine pancreatic arteries appear to lack vasorelaxant P2Y(2) and P2Y(4) receptors.


Asunto(s)
Arterias/metabolismo , Endotelio Vascular/metabolismo , Páncreas/metabolismo , Nucleótidos de Pirimidina/metabolismo , Receptores Purinérgicos/metabolismo , Animales , Endotelio Vascular/efectos de los fármacos , Páncreas/irrigación sanguínea , Porcinos , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Vasodilatadores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA