Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Biol Chem ; 299(8): 105011, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37414150

RESUMEN

The pentose phosphate pathway (PPP) is critical for anabolism and biomass production. Here we show that the essential function of PPP in yeast is the synthesis of phosphoribosyl pyrophosphate (PRPP) catalyzed by PRPP-synthetase. Using combinations of yeast mutants, we found that a mildly decreased synthesis of PRPP affects biomass production, resulting in reduced cell size, while a more severe decrease ends up affecting yeast doubling time. We establish that it is PRPP itself that is limiting in invalid PRPP-synthetase mutants and that the resulting metabolic and growth defect can be bypassed by proper supplementation of the medium with ribose-containing precursors or by the expression of bacterial or human PRPP-synthetase. In addition, using documented pathologic human hyperactive forms of PRPP-synthetase, we show that intracellular PRPP as well as its derived products can be increased in both human and yeast cells, and we describe the ensuing metabolic and physiological consequences. Finally, we found that PRPP consumption appears to take place "on demand" by the various PRPP-utilizing pathways, as shown by blocking or increasing the flux in specific PRPP-consuming metabolic routes. Overall, our work reveals important similarities between human and yeast for both synthesis and consumption of PRPP.


Asunto(s)
Fosforribosil Pirofosfato , Saccharomyces cerevisiae , Humanos , Saccharomyces cerevisiae/genética , Bacterias , Vía de Pentosa Fosfato , Ligasas
2.
J Biol Chem ; 294(3): 805-815, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30478173

RESUMEN

5-Aminoimidazole-4-carboxamide 1-ß-d-ribofuranoside (AICAR, or acadesine) is a precursor of the monophosphate derivative 5-amino-4-imidazole carboxamide ribonucleoside 5'-phosphate (ZMP), an intermediate in de novo purine biosynthesis. AICAR proved to have promising anti-proliferative properties, although the molecular basis of its toxicity is poorly understood. To exert cytotoxicity, AICAR needs to be metabolized, but the AICAR-derived toxic metabolite was not identified. Here, we show that ZMP is the major toxic derivative of AICAR in yeast and establish that its metabolization to succinyl-ZMP, ZDP, or ZTP (di- and triphosphate derivatives of AICAR) strongly reduced its toxicity. Affinity chromatography identified 74 ZMP-binding proteins, including 41 that were found neither as AMP nor as AICAR or succinyl-ZMP binders. Overexpression of karyopherin-ß Kap123, one of the ZMP-specific binders, partially rescued AICAR toxicity. Quantitative proteomic analyses revealed 57 proteins significantly less abundant on nuclei-enriched fractions from AICAR-fed cells, this effect being compensated by overexpression of KAP123 for 15 of them. These results reveal nuclear protein trafficking as a function affected by AICAR.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proteómica , Ribonucleótidos , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Aminoimidazol Carboxamida/farmacocinética , Aminoimidazol Carboxamida/farmacología , Núcleo Celular/química , Núcleo Celular/genética , Cromatografía de Afinidad , Ribonucleótidos/farmacocinética , Ribonucleótidos/farmacología , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
3.
J Biol Chem ; 294(32): 11980-11991, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31160323

RESUMEN

The reversible adenine phosphoribosyltransferase enzyme (APRT) is essential for purine homeostasis in prokaryotes and eukaryotes. In humans, APRT (hAPRT) is the only enzyme known to produce AMP in cells from dietary adenine. APRT can also process adenine analogs, which are involved in plant development or neuronal homeostasis. However, the molecular mechanism underlying substrate specificity of APRT and catalysis in both directions of the reaction remains poorly understood. Here we present the crystal structures of hAPRT complexed to three cellular nucleotide analogs (hypoxanthine, IMP, and GMP) that we compare with the phosphate-bound enzyme. We established that binding to hAPRT is substrate shape-specific in the forward reaction, whereas it is base-specific in the reverse reaction. Furthermore, a quantum mechanics/molecular mechanics (QM/MM) analysis suggests that the forward reaction is mainly a nucleophilic substitution of type 2 (SN2) with a mix of SN1-type molecular mechanism. Based on our structural analysis, a magnesium-assisted SN2-type mechanism would be involved in the reverse reaction. These results provide a framework for understanding the molecular mechanism and substrate discrimination in both directions by APRTs. This knowledge can play an instrumental role in the design of inhibitors, such as antiparasitic agents, or adenine-based substrates.


Asunto(s)
Adenina Fosforribosiltransferasa/metabolismo , Adenina/química , Adenina/metabolismo , Adenina Fosforribosiltransferasa/química , Biocatálisis , Cristalografía por Rayos X , Humanos , Cinética , Modelos Moleculares , Estructura Terciaria de Proteína , Teoría Cuántica , Especificidad por Sustrato
4.
Curr Genet ; 66(6): 1163-1177, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32780163

RESUMEN

Because metabolism is a complex balanced process involving multiple enzymes, understanding how organisms compensate for transient or permanent metabolic imbalance is a challenging task that can be more easily achieved in simpler unicellular organisms. The metabolic balance results not only from the combination of individual enzymatic properties, regulation of enzyme abundance, but also from the architecture of the metabolic network offering multiple interconversion alternatives. Although metabolic networks are generally highly resilient to perturbations, metabolic imbalance resulting from enzymatic defect and specific environmental conditions can be designed experimentally and studied. Starting with a double amd1 aah1 mutant that severely and conditionally affects yeast growth, we carefully characterized the metabolic shuffle associated with this defect. We established that the GTP decrease resulting in an adenylic/guanylic nucleotide imbalance was responsible for the growth defect. Identification of several gene dosage suppressors revealed that TAT1, encoding an amino acid transporter, is a robust suppressor of the amd1 aah1 growth defect. We show that TAT1 suppression occurs through replenishment of the GTP pool in a process requiring the histidine biosynthesis pathway. Importantly, we establish that a tat1 mutant exhibits synthetic sickness when combined with an amd1 mutant and that both components of this synthetic phenotype can be suppressed by specific gene dosage suppressors. Together our data point to a strong phenotypic connection between amino acid uptake and GTP synthesis, a connection that could open perspectives for future treatment of related human defects, previously reported as etiologically highly conserved.


Asunto(s)
AMP Desaminasa/genética , Sistemas de Transporte de Aminoácidos/genética , Aminohidrolasas/genética , Nucleósidos de Purina/genética , Proteínas de Saccharomyces cerevisiae/genética , Guanosina Trifosfato/genética , Humanos , Nucleótidos/genética , Fenotipo , Saccharomyces cerevisiae/genética
5.
Curr Genet ; 64(6): 1275-1286, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29721631

RESUMEN

AICAR is the precursor of ZMP, a metabolite with antiproliferative properties in yeast and human. We aim at understanding how AICAR (and its active form ZMP) affects essential cellular processes. In this work, we found that ZMP accumulation is synthetic lethal with a hypomorphic allele of the ubiquitin-activating enzyme Uba1. A search for gene-dosage suppressors revealed that ubiquitin overexpression was sufficient to restore growth of the uba1 mutant upon AICAR treatment, suggesting that the ubiquitin pool is critical for cells to cope with AICAR. Accordingly, two mutants with constitutive low ubiquitin, ubp6 and doa1, were highly sensitive to AICAR, a phenotype that could be suppressed by ubiquitin overexpression. We established, by genetic means, that these new AICAR-sensitive mutants act in a different pathway from the rad6/bre1 mutants which were previously reported as sensitive to AICAR (Albrecht et al., Genetics 204:1447-1460, 2016). Two ubiquitin-conjugating enzymes (Ubc4 and Cdc34) and a ubiquitin ligase (Cdc4) were found to contribute to the ability of cells to cope with ZMP. This study illustrates the complexity of chemo-genetic interactions and shows how genetic analyses allow deciphering the implicated pathways, the individual gene effects, and their combined phenotypic contribution. Based on additivity and suppression patterns, we conclude that AICAR treatment shows synthetic interactions with distinct branches of the yeast ubiquitin pathway.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Ribonucleótidos/farmacología , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Ubiquitina , Ubiquitinación/efectos de los fármacos , Aminoimidazol Carboxamida/farmacología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitinación/genética
6.
J Biol Chem ; 290(39): 23947-59, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26283791

RESUMEN

5-Aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside monophosphate (AICAR) is a natural metabolite with potent anti-proliferative and low energy mimetic properties. At high concentration, AICAR is toxic for yeast and mammalian cells, but the molecular basis of this toxicity is poorly understood. Here, we report the identification of yeast purine salvage pathway mutants that are synthetically lethal with AICAR accumulation. Genetic suppression revealed that this synthetic lethality is in part due to low expression of adenine phosphoribosyl transferase under high AICAR conditions. In addition, metabolite profiling points to the AICAR/NTP balance as crucial for optimal utilization of glucose as a carbon source. Indeed, we found that AICAR toxicity in yeast and human cells is alleviated when glucose is replaced by an alternative carbon source. Together, our metabolic analyses unveil the AICAR/NTP balance as a major factor of AICAR antiproliferative effects.


Asunto(s)
Adenina Fosforribosiltransferasa/antagonistas & inhibidores , Aminoimidazol Carboxamida/análogos & derivados , Proliferación Celular/efectos de los fármacos , Nucleótidos/metabolismo , Ribonucleótidos/farmacología , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Saccharomyces cerevisiae/metabolismo , Adenina Fosforribosiltransferasa/genética , Adenina Fosforribosiltransferasa/metabolismo , Aminoimidazol Carboxamida/farmacología , Línea Celular , Proliferación Celular/genética , Humanos , Nucleótidos/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
7.
J Biol Chem ; 289(24): 16844-54, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24778186

RESUMEN

5-Aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAr) is the precursor of the active monophosphate form (AICAR), a small molecule with potent anti-proliferative and low energy mimetic properties. The molecular bases for AICAR toxicity at the cellular level are poorly understood. Here, we report the isolation and characterization of several yeast AICAr-hypersensitive mutants. Identification of the cognate genes allowed us to establish that thiamine transporters Thi7 and Thi72 can efficiently take up AICAr under conditions where they are overexpressed. We establish that, under standard growth conditions, Nrt1, the nicotinamide riboside carrier, is the major AICAr transporter in yeast. A study of AICAR accumulation in human cells revealed substantial disparities among cell lines and confirmed that AICAr enters cells via purine nucleoside transporters. Together, our results point to significant differences between yeast and human cells for both AICAr uptake and AICAR accumulation.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Proteínas de Transporte de Membrana/metabolismo , Ribonucleótidos/farmacología , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Aminoimidazol Carboxamida/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Humanos , Proteínas de Transporte de Membrana/genética , Ratones , Mutación , Proteínas de Transporte de Nucleósidos/genética , Proteínas de Transporte de Nucleósidos/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Tiamina/metabolismo
8.
Curr Genet ; 61(4): 633-40, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25893566

RESUMEN

Previous genetic analyses showed phenotypic interactions between 5-amino-4-imidazole carboxamide ribonucleotide 5'-phosphate (AICAR) produced from the purine and histidine pathways and methionine biosynthesis. Here, we revisited the effect of AICAR on methionine requirement due to AICAR accumulation in the presence of the fau1 mutation invalidating folinic acid remobilization. We found that this methionine auxotrophy could be suppressed by overexpression of the methionine synthase Met6 or by deletion of the serine hydroxymethyltransferase gene SHM2. We propose that in a fau1 background, AICAR, by stimulating the transcriptional expression of SHM2, leads to a folinic acid accumulation inhibiting methionine synthesis by Met6. In addition, we uncovered a new methionine auxotrophy for the ade3 bas1 double mutant that can be rescued by overexpressing the SHM2 gene. We propose that methionine auxotrophy in this mutant is the result of a competition for 5,10-methylenetetrahydrofolate between methionine and deoxythymidine monophosphate synthesis. Altogether, our data show intricate genetic interactions between one-carbon units, purine and methionine metabolism through fine-tuning of serine hydroxymethyltransferase by AICAR and the transcription factor Bas1.


Asunto(s)
Ácido Fólico/metabolismo , Regulación Fúngica de la Expresión Génica , Glicina Hidroximetiltransferasa/metabolismo , Metionina/metabolismo , Purinas/metabolismo , Saccharomyces cerevisiae/genética , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/metabolismo , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Glicina Hidroximetiltransferasa/genética , Histidina/metabolismo , Leucovorina/metabolismo , Mutación , Ribonucleótidos/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Tetrahidrofolatos/metabolismo , Timidina/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Activación Transcripcional
9.
Cells ; 12(19)2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37830593

RESUMEN

Purines are required for fundamental biological processes and alterations in their metabolism lead to severe genetic diseases associated with developmental defects whose etiology remains unclear. Here, we studied the developmental requirements for purine metabolism using the amphibian Xenopus laevis as a vertebrate model. We provide the first functional characterization of purine pathway genes and show that these genes are mainly expressed in nervous and muscular embryonic tissues. Morphants were generated to decipher the functions of these genes, with a focus on the adenylosuccinate lyase (ADSL), which is an enzyme required for both salvage and de novo purine pathways. adsl.L knockdown led to a severe reduction in the expression of the myogenic regulatory factors (MRFs: Myod1, Myf5 and Myogenin), thus resulting in defects in somite formation and, at later stages, the development and/or migration of both craniofacial and hypaxial muscle progenitors. The reduced expressions of hprt1.L and ppat, which are two genes specific to the salvage and de novo pathways, respectively, resulted in similar alterations. In conclusion, our data show for the first time that de novo and recycling purine pathways are essential for myogenesis and highlight new mechanisms in the regulation of MRF gene expression.


Asunto(s)
Músculo Esquelético , Purinas , Animales , Xenopus laevis/genética , Músculo Esquelético/metabolismo , Purinas/metabolismo , Desarrollo de Músculos/genética
10.
J Biol Chem ; 286(35): 30994-31002, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21757731

RESUMEN

5-Amino-4-imidazolecarboxamide ribonucleotide 5'-phosphate (AICAR) is a monophosphate metabolic intermediate of the de novo purine synthesis pathway that has highly promising metabolic and antiproliferative properties. Yeast mutants unable to metabolize AICAR are auxotroph for histidine. A screening for suppressors of this phenotype identified recessive and dominant mutants that result in lowering the intracellular AICAR concentration. The recessive mutants affect the adenosine kinase, which is shown here to catalyze the phosphorylation of AICAR riboside in yeast. The dominant mutants strongly enhance the capacity of the alkaline phosphatase Pho13 to dephosphorylate 5-amino-4-imidazole N-succinocarboxamide ribonucleotide 5'-phosphate(SAICAR) into its non-toxic riboside form. By combining these mutants with transcriptomics and metabolomics analyses, we establish that in yeast responses to AICAR and SAICAR are clearly linked to the concentration of the monophosphate forms, whereas the derived nucleoside moieties have no effect even at high intracellular concentration. Finally, we show that AICAR/SAICAR concentrations vary under physiological conditions known to modulate transcription of the purine and phosphate pathway genes.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Genes Fúngicos , Mutación , Purinas/química , Ribonucleótidos/genética , Fosfatasa Alcalina/metabolismo , Catálisis , Cromatografía Liquida/métodos , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Genes Dominantes , Genes Recesivos , Modelos Químicos , Saccharomyces cerevisiae/genética , Especificidad de la Especie , Transcripción Genética
11.
Elife ; 82019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30860478

RESUMEN

Metabolism is a highly integrated process resulting in energy and biomass production. While individual metabolic routes are well characterized, the mechanisms ensuring crosstalk between pathways are poorly described, although they are crucial for homeostasis. Here, we establish a co-regulation of purine and pyridine metabolism in response to external adenine through two separable mechanisms. First, adenine depletion promotes transcriptional upregulation of the de novo NAD+ biosynthesis genes by a mechanism requiring the key-purine intermediates ZMP/SZMP and the Bas1/Pho2 transcription factors. Second, adenine supplementation favors the pyridine salvage route resulting in an ATP-dependent increase of intracellular NAD+. This control operates at the level of the nicotinic acid mononucleotide adenylyl-transferase Nma1 and can be bypassed by overexpressing this enzyme. Therefore, in yeast, pyridine metabolism is under the dual control of ZMP/SZMP and ATP, revealing a much wider regulatory role for these intermediate metabolites in an integrated biosynthesis network.


Asunto(s)
Proteínas Fúngicas/metabolismo , Regulación Neoplásica de la Expresión Génica , NAD/biosíntesis , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Purinas/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Adenina/química , Adenosina Trifosfato/química , Biomasa , Cromatografía Liquida , Genotipo , Proteínas de Homeodominio/metabolismo , Homeostasis , Niacina/química , Nicotinamida-Nucleótido Adenililtransferasa/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
12.
Genetics ; 211(4): 1297-1313, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30700528

RESUMEN

Purine homeostasis is ensured through a metabolic network widely conserved from prokaryotes to humans. Purines can either be synthesized de novo, reused, or produced by interconversion of extant metabolites using the so-called recycling pathway. Although thoroughly characterized in microorganisms, such as yeast or bacteria, little is known about regulation of the purine biosynthesis network in metazoans. In humans, several diseases are linked to purine metabolism through as yet poorly understood etiologies. Particularly, the deficiency in adenylosuccinate lyase (ADSL)-an enzyme involved both in the purine de novo and recycling pathways-causes severe muscular and neuronal symptoms. In order to address the mechanisms underlying this deficiency, we established Caenorhabditis elegans as a metazoan model organism to study purine metabolism, while focusing on ADSL. We show that the purine biosynthesis network is functionally conserved in C. elegans Moreover, adsl-1 (the gene encoding ADSL in C. elegans) is required for developmental timing, germline stem cell maintenance and muscle integrity. Importantly, these traits are not affected when solely the de novo pathway is abolished, and we present evidence that germline maintenance is linked specifically to ADSL activity in the recycling pathway. Hence, our results allow developmental and tissue specific phenotypes to be ascribed to separable steps of the purine metabolic network in an animal model.


Asunto(s)
Adenilosuccinato Liasa/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Células Germinativas/metabolismo , Homeostasis , Músculo Esquelético/metabolismo , Purinas/metabolismo , Adenilosuccinato Liasa/genética , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Células Germinativas/citología
13.
Cell Chem Biol ; 25(6): 666-676.e4, 2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29576532

RESUMEN

Phosphoribosyltransferases catalyze the displacement of a PRPP α-1'-pyrophosphate to a nitrogen-containing nucleobase. How they control the balance of substrates/products binding and activities is poorly understood. Here, we investigated the human adenine phosphoribosyltransferase (hAPRT) that produces AMP in the purine salvage pathway. We show that a single oxygen atom from the Tyr105 side chain is responsible for selecting the active conformation of the 12 amino acid long catalytic loop. Using in vitro, cellular, and in crystallo approaches, we demonstrated that Tyr105 is key for the fine-tuning of the kinetic activity efficiencies of the forward and reverse reactions. Together, our results reveal an evolutionary pressure on the strictly conserved Tyr105 and on the dynamic motion of the flexible loop in phosphoribosyltransferases that is essential for purine biosynthesis in cells. These data also provide the framework for designing novel adenine derivatives that could modulate, through hAPRT, diseases-involved cellular pathways.


Asunto(s)
Adenina Fosforribosiltransferasa/metabolismo , Adenina Fosforribosiltransferasa/química , Adenina Fosforribosiltransferasa/aislamiento & purificación , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Conformación Proteica
14.
BMC Biochem ; 4: 4, 2003 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-12735798

RESUMEN

BACKGROUND: The purine salvage enzyme inosine 5'-monophosphate (IMP)-specific 5'-nucleotidase catalyzes degradation of IMP to inosine. Although this enzymatic activity has been purified and characterized in Saccharomyces cerevisiae, the gene encoding IMP 5'-nucleotidase had not been identified. RESULTS: Mass spectrometry analysis of several peptides of this enzyme purified from yeast allowed identification of the corresponding gene as YOR155c, an open reading frame of unknown function, renamed ISN1. The deduced Isn1p sequence was clearly not homologous to 5'-nucleotidases from other species. However, significant similarities to Isn1p were found in proteins of unknown function from Neurospora crassa, Plasmodium falciparum and several yeast species. Knock-out of ISN1 resulted in the total loss of IMP-specific 5'-nucleotidase activity, thus confirming that the ISN1 gene indeed encodes the enzymatic activity purified from yeast. In vivo studies revealed that, when IMP is overproduced through constitutive activation of the IMP de novo synthesis pathway, ISN1 is required for excretion of inosine and hypoxanthine in the medium. CONCLUSION: We have identified a new yeast gene, ISN1 (YOR155c), as encoding IMP-specific 5'-nucleotidase activity. The ISN1 gene defines a new type of 5'-nucleotidase which was demonstrated to be functional in vivo.


Asunto(s)
5'-Nucleotidasa/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , 5'-Nucleotidasa/clasificación , Secuencia de Aminoácidos , Genes Fúngicos , Datos de Secuencia Molecular , Monoéster Fosfórico Hidrolasas/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/clasificación , Proteínas de Saccharomyces cerevisiae/genética , Homología de Secuencia de Aminoácido
15.
Genetics ; 183(2): 529-38, 1SI-7SI, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19635936

RESUMEN

Coordinating homeostasis of multiple metabolites is a major task for living organisms, and complex interconversion pathways contribute to achieving the proper balance of metabolites. AMP deaminase (AMPD) is such an interconversion enzyme that allows IMP synthesis from AMP. In this article, we show that, under specific conditions, lack of AMPD activity impairs growth. Under these conditions, we found that the intracellular guanylic nucleotide pool was severely affected. In vivo studies of two AMPD homologs, Yjl070p and Ybr284p, indicate that these proteins have no detectable AMP, adenosine, or adenine deaminase activity; we show that overexpression of YJL070c instead mimics a loss of AMPD function. Expression of the yeast transcriptome was monitored in a AMPD-deficient mutant in a strain overexpressing YJL070c and in cells treated with the immunosuppressive drug mycophenolic acid, three conditions that lead to severe depletion of the guanylic nucleotide pool. These three conditions resulted in the up- or downregulation of multiple transcripts, 244 of which are common to at least two conditions and 71 to all three conditions. These transcriptome results, combined with specific mutant analysis, point to threonine metabolism as exquisitely sensitive to the purine nucleotide balance.


Asunto(s)
AMP Desaminasa/metabolismo , Nucleótidos de Purina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , AMP Desaminasa/genética , Vías Biosintéticas/efectos de los fármacos , División Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación Fúngica de la Expresión Génica/genética , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Inosina Monofosfato/biosíntesis , Inosina Monofosfato/metabolismo , Mutación , Ácido Micofenólico/farmacología , Fenotipo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Treonina/metabolismo
16.
Yeast ; 21(16): 1359-63, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15565584

RESUMEN

Purine salvage is a complex pathway allowing a correct balance between adenylic and guanylic derivatives. In this paper, we show that GUD1 (YDL238c) encodes guanine deaminase, a catabolic enzyme producing xanthine and ammonia from guanine. Importantly, Gud1p activity was higher during post-diauxic growth, suggesting that a decrease of the guanylic nucleotide pool could be required when cells shift from proliferation to quiescence.


Asunto(s)
Genes Fúngicos , Guanina Desaminasa/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Guanina Desaminasa/biosíntesis , Guanina Desaminasa/química , Datos de Secuencia Molecular , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/biosíntesis , Alineación de Secuencia
17.
J Biol Chem ; 277(30): 27036-44, 2002 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-12016207

RESUMEN

The immunosuppressive drug mycophenolic acid (MPA) is a potent and specific inhibitor of IMP dehydrogenase, the first committed step of GMP synthesis. A screen for yeast genes affecting MPA sensitivity, when overexpressed, allowed us to identify two genes, IMD2 and TPO1, encoding a homologue of IMP dehydrogenase and a vacuolar pump, respectively. In parallel, 4787 yeast strains, each carrying an identified knock-out mutation, were tested for growth in the presence of MPA, allowing identification of 100 new genes affecting MPA resistance when disrupted. Disturbance of several cellular processes, such as ergosterol biosynthesis, vacuole biogenesis, or glycosylation impaired the natural capacity of yeast to resist MPA, although most of the highly sensitive mutants affected the transcription machinery (19 mutants). Expression of TPO1 and/or IMD2 was strongly affected in 16 such transcription mutants suggesting that low expression of these genes could contribute to MPA sensitivity. Interestingly, the spt3, spt8, and spt20 mutants behaved differently than other Spt-Ada-Gcn5-acetyltransferase (SAGA) mutants. Indeed, in these three mutants, as in previously characterized transcription elongation mutants, IMD2 expression was only affected in the presence of MPA, thus suggesting a possible role for some SAGA subunits in transcription elongation.


Asunto(s)
Resistencia a Medicamentos/genética , Inmunosupresores/farmacología , Mutación , Ácido Micofenólico/farmacología , Northern Blotting , Inhibidores Enzimáticos/farmacología , Modelos Genéticos , Sistemas de Lectura Abierta , Fenotipo , Plásmidos/metabolismo , Saccharomyces cerevisiae/genética , Transcripción Genética , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA