Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
J Biol Chem ; 294(3): 805-815, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30478173

RESUMO

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.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Proteômica , Ribonucleotídeos , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Aminoimidazol Carboxamida/farmacocinética , Aminoimidazol Carboxamida/farmacologia , Núcleo Celular/química , Núcleo Celular/genética , Cromatografia de Afinidade , Ribonucleotídeos/farmacocinética , Ribonucleotídeos/farmacologia , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
2.
J Biol Chem ; 294(31): 11920-11933, 2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-31201273

RESUMO

Human guanylate kinase (hGMPK) is the only known enzyme responsible for cellular GDP production, making it essential for cellular viability and proliferation. Moreover, hGMPK has been assigned a critical role in metabolic activation of antiviral and antineoplastic nucleoside-analog prodrugs. Given that hGMPK is indispensable for producing the nucleotide building blocks of DNA, RNA, and cGMP and that cancer cells possess elevated GTP levels, it is surprising that a detailed structural and functional characterization of hGMPK is lacking. Here, we present the first high-resolution structure of hGMPK in the apo form, determined with NMR spectroscopy. The structure revealed that hGMPK consists of three distinct regions designated as the LID, GMP-binding (GMP-BD), and CORE domains and is in an open configuration that is nucleotide binding-competent. We also demonstrate that nonsynonymous single-nucleotide variants (nsSNVs) of the hGMPK CORE domain distant from the nucleotide-binding site of this domain modulate enzymatic activity without significantly affecting hGMPK's structure. Finally, we show that knocking down the hGMPK gene in lung adenocarcinoma cell lines decreases cellular viability, proliferation, and clonogenic potential while not altering the proliferation of immortalized, noncancerous human peripheral airway cells. Taken together, our results provide an important step toward establishing hGMPK as a potential biomolecular target, from both an orthosteric (ligand-binding sites) and allosteric (location of CORE domain-located nsSNVs) standpoint.


Assuntos
Guanilato Quinases/metabolismo , Regulação Alostérica , Animais , Linhagem Celular Tumoral , Cristalografia por Raios X , Guanilato Quinases/química , Guanilato Quinases/genética , Humanos , Cinética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Mutagênese Sítio-Dirigida , Ressonância Magnética Nuclear Biomolecular , Estrutura Terciária de Proteína , Interferência de RNA , RNA Interferente Pequeno , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação
3.
Eur Biophys J ; 45(1): 81-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26446352

RESUMO

Bio-catalysis is the outcome of a subtle interplay between internal motions in enzymes and chemical kinetics. Small-angle X-ray scattering (SAXS) investigation of an enzyme's internal motions during catalysis offers an integral view of the protein's structural plasticity, dynamics, and function, which is useful for understanding allosteric effects and developing novel medicines. Guanylate kinase (GMPK) is an essential enzyme involved in the guanine nucleotide metabolism of unicellular and multicellular organisms. It is also required for the intracellular activation of numerous antiviral and anticancer purine nucleoside analog prodrugs. Catalytically active recombinant human GMPK (hGMPK) was purified for the first time and changes in the size and shape of open/closed hGMPK were tracked by SAXS. The binding of substrates (GMP + AMPPNP or Ap5G or GMP + ADP) resulted in the compaction of size and shape of hGMPK. The structural changes between open and completely closed hGMPK conformation were confirmed by observing differences in the hGMPK secondary structures with circular dichroism spectroscopy.


Assuntos
Domínio Catalítico , Guanilato Ciclase/química , Simulação de Dinâmica Molecular , Sequência de Aminoácidos , Humanos , Dados de Sequência Molecular , Espalhamento a Baixo Ângulo , Difração de Raios X
4.
J Biol Chem ; 289(19): 12962-75, 2014 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-24657844

RESUMO

The structural and functional characterization of human enzymes that are of potential medical and therapeutic interest is of prime significance for translational research. One of the most notable examples of a therapeutic enzyme is L-asparaginase, which has been established as an antileukemic protein drug for more than four decades. Up until now, only bacterial enzymes have been used in therapy despite a plethora of undesired side effects mainly attributed to the bacterial origins of these enzymes. Therefore, the replacement of the currently approved bacterial drugs by human homologs aiming at the elimination of adverse effects is of great importance. Recently, we structurally and biochemically characterized the enzyme human L-asparaginase 3 (hASNase3), which possesses L-asparaginase activity and belongs to the N-terminal nucleophile superfamily of enzymes. Inspired by the necessity for the development of a protein drug of human origin, in the present study, we focused on the characterization of another human L-asparaginase, termed hASNase1. This bacterial-type cytoplasmic L-asparaginase resides in the N-terminal subdomain of an overall 573-residue protein previously reported to function as a lysophospholipase. Our kinetic, mutagenesis, structural modeling, and fluorescence labeling data highlight allosteric features of hASNase1 that are similar to those of its Escherichia coli homolog, EcASNase1. Differential scanning fluorometry and urea denaturation experiments demonstrate the impact of particular mutations on the structural and functional integrity of the L-asparaginase domain and provide a direct comparison of sites critical for the conformational stability of the human and E. coli enzymes.


Assuntos
Asparaginase/química , Asparagina/química , Lisofosfolipase/química , Modelos Moleculares , Regulação Alostérica/fisiologia , Asparaginase/genética , Asparaginase/metabolismo , Asparagina/genética , Asparagina/metabolismo , Estabilidade Enzimática , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Lisofosfolipase/genética , Lisofosfolipase/metabolismo , Estrutura Terciária de Proteína , Homologia Estrutural de Proteína
5.
Small ; 11(43): 5844-50, 2015 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-26395754

RESUMO

A photo-electrochemical sensor for the specific detection of guanosine monophosphate (GMP) is demonstrated, based on three enzymes combined in a coupled reaction assay. The first reaction involves the adenosine triphosphate (ATP)-dependent conversion of GMP to guanosine diphosphate (GDP) by guanylate kinase, which warrants substrate specificity. The reaction products ADP and GDPare co-substrates for the enzymatic conversion of phosphoenolpyruvate to pyruvate in a second reaction mediated by pyruvate kinase. Pyruvate in turn is the co-substrate for lactate dehydrogenase that generates lactate via oxidation of nicotinamide adenine dinucleotide (reduced form) NADH to NAD(+). This third enzymatic reaction is electrochemically detected. For this purpose a CdS/ZnS quantum dot (QD) electrode is illuminated and the photocurrent response under fixed potential conditions is evaluated. The sequential enzyme reactions are first evaluated in solution. Subsequently, a sensor for GMP is constructed using polyelectrolytes for enzyme immobilization.


Assuntos
Técnicas Biossensoriais/instrumentação , Condutometria/instrumentação , Guanosina Monofosfato/análise , L-Lactato Desidrogenase/química , Pontos Quânticos , Espectrometria de Fluorescência/instrumentação , Compostos de Cádmio/química , Enzimas Imobilizadas , Desenho de Equipamento , Análise de Falha de Equipamento , Microeletrodos , Compostos de Selênio/química , Compostos de Zinco/química
6.
Anal Biochem ; 445: 20-3, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24113285

RESUMO

We report on the development of a sensitive real-time assay for monitoring the activity of L-asparaginase that hydrolyzes L-asparagine to L-aspartate and ammonia. In this method, L-aspartate is oxidized by L-aspartate oxidase to iminoaspartate and hydrogen peroxide (H2O2), and in the detection step horseradish peroxidase uses H2O2 to convert the colorless, nonfluorescent reagent Amplex Red to the red-colored and highly fluorescent product resorufin. The assay was validated in both the absorbance and the fluorescence modes. We show that, due to its high sensitivity and substrate selectivity, this assay can be used to measure enzymatic activity in human serum containing L-asparaginase.


Assuntos
Asparaginase/metabolismo , Fluorometria , Oxazinas/química , Espectrometria de Fluorescência , Ensaios Enzimáticos , Humanos , Oxazinas/metabolismo , Oxirredução , Especificidade por Substrato
7.
Protein Expr Purif ; 93: 1-10, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24157738

RESUMO

l-asparaginases hydrolyze l-asparagine to l-aspartic acid and ammonia. Enzymes of bacterial origin are used as therapeutic agents for the treatment of acute lymphoblastic leukemia. Recently, the structure of a human homolog, hASNase3, which possesses l-asparaginase activity, was solved setting the basis for the development of an anti-leukemic protein drug of human origin. Being an N-terminal hydrolase, hASNase3 undergoes intramolecular self-cleavage generating two protomers (subunits α and ß) which remain non-covalently associated and constitute the catalytically active form of the enzyme. However, recombinant expression of full-length hASNase3 in Escherichiacoli results in only partial processing towards the active enzyme. We developed a co-expression system for the two subunits that allowed production of the ß-subunit complexed to the α-subunit such that the N-terminal methionine is removed by endogenous methionine aminopeptidase to expose the catalytically essential threonine residue at the N-terminus of the ß-subunit. The enzyme produced by this co-expression strategy is fully active, thus obviating the necessity of self-activation by slow autoproteolytic cleavage.

8.
Biomacromolecules ; 14(12): 4398-406, 2013 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24144040

RESUMO

The present study focuses on the formation of microcapsules containing catalytically active L-asparaginase (L-ASNase), a protein drug of high value in antileukemic therapy. We make use of the layer-by-layer (LbL) technique to coat protein-loaded calcium carbonate (CaCO3) particles with two or three poly dextran/poly-L-arginine-based bilayers. To achieve high loading efficiency, the CaCO3 template was generated by coprecipitation with the enzyme. After assembly of the polymer shell, the CaCO3 core material was dissolved under mild conditions by dialysis against 20 mM EDTA. Biochemical stability of the encapsulated L-asparaginase was analyzed by treating the capsules with the proteases trypsin and thrombin, which are known to degrade and inactivate the enzyme during leukemia treatment, allowing us to test for resistance against proteolysis by physiologically relevant proteases through measurement of residual l-asparaginase activities. In addition, the thermal stability, the stability at the physiological temperature, and the long-term storage stability of the encapsulated enzyme were investigated. We show that encapsulation of l-asparaginase remarkably improves both proteolytic resistance and thermal inactivation at 37 °C, which could considerably prolong the enzyme's in vivo half-life during application in acute lymphoblastic leukemia (ALL). Importantly, the use of low EDTA concentrations for the dissolution of CaCO3 by dialysis could be a general approach in cases where the activity of sensitive biomacromolecules is inhibited, or even irreversibly damaged, when standard protocols for fabrication of such LbL microcapsules are used. Encapsulated and free enzyme showed similar efficacies in driving leukemic cells to apoptosis.


Assuntos
Asparaginase/química , Portadores de Fármacos/química , Proteínas de Escherichia coli/química , Polímeros/química , Proteínas de Saccharomyces cerevisiae/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Asparaginase/farmacologia , Materiais Biocompatíveis/química , Carbonato de Cálcio/química , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Eletrólitos/química , Estabilidade Enzimática , Escherichia coli/enzimologia , Proteínas de Escherichia coli/farmacologia , Humanos , Saccharomyces cerevisiae/enzimologia , Proteínas de Saccharomyces cerevisiae/farmacologia
9.
Biochemistry ; 51(34): 6816-26, 2012 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-22861376

RESUMO

Asparaginases catalyze the hydrolysis of the amino acid asparagine to aspartate and ammonia. Bacterial asparaginases are used in cancer chemotherapy to deplete asparagine from the blood, because several hematological malignancies depend on extracellular asparagine for growth. To avoid the immune response against the bacterial enzymes, it would be beneficial to replace them with human asparaginases. However, unlike the bacterial asparaginases, the human enzymes have a millimolar K(m) value for asparagine, making them inefficient in depleting the amino acid from blood. To facilitate the development of human variants suitable for therapeutic use, we determined the structure of human l-asparaginase (hASNase3). This asparaginase is an N-terminal nucleophile (Ntn) family member that requires autocleavage between Gly167 and Thr168 to become catalytically competent. For most Ntn hydrolases, this autoproteolytic activation occurs efficiently. In contrast, hASNas3 is relatively stable in its uncleaved state, and this allowed us to observe the structure of the enzyme prior to cleavage. To determine the structure of the cleaved state, we exploited our discovery that the free amino acid glycine promotes complete cleavage of hASNase3. Both enzyme states were elucidated in the absence and presence of the product aspartate. Together, these structures provide insight into the conformational changes required for cleavage and the precise enzyme-substrate interactions. The new understanding of hASNase3 will serve to guide the design of variants that possess a decreased K(m) value for asparagine, making the human enzyme a suitable replacement for the bacterial asparaginases in cancer therapy.


Assuntos
Asparaginase/química , Asparaginase/metabolismo , Asparagina/metabolismo , Sequência de Aminoácidos , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Asparaginase/genética , Sítios de Ligação , Biocatálise , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Alinhamento de Sequência , Especificidade por Substrato
10.
Biochemistry ; 50(14): 2870-80, 2011 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21351740

RESUMO

Deoxycytidine kinase (dCK) uses either ATP or UTP as a phosphoryl donor to catalyze the phosphorylation of nucleoside acceptors. The kinetic properties of human dCK are modulated in vivo by phosphorylation of serine 74. This residue is a part of the insert region and is distant from the active site. Replacing the serine with a glutamic acid (S74E variant) can mimic phosphorylation of Ser74. To understand how phosphorylation affects the catalytic properties of dCK, we examined the S74E variant of dCK both structurally and kinetically. We observe that the presence of a glutamic acid at position 74 favors the adoption by the enzyme of the open conformation. Glu74 stabilizes the open conformation by directly interacting with the indole side chain of Trp58, a residue that is in the proximity of the base of the nucleoside substrate. The open dCK conformation is competent for the binding of nucleoside but not for phosphoryl transfer. In contrast, the closed conformation is competent for phosphoryl transfer but not for product release. Thus, dCK must make the transition between the open and closed states during the catalytic cycle. We propose a reaction scheme for dCK that incorporates the transition between the open and closed states, and this serves to rationalize the observed kinetic differences between wild-type dCK and the S74E variant.


Assuntos
Desoxicitidina Quinase/química , Desoxicitidina Quinase/metabolismo , Nucleosídeos/metabolismo , Conformação Proteica , Serina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação/genética , Biocatálise , Desoxicitidina Quinase/genética , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Serina/genética
11.
Biochemistry ; 49(31): 6784-90, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20614893

RESUMO

The physiological role of human deoxycytidine kinase (dCK) is to phosphorylate deoxynucleosides required for DNA synthesis, with the exception of thymidine. Previous structural analysis of dCK implicated steric factors, specifically the thymine methyl group at the 5-position, that prevent thymidine phosphorylation by dCK. This hypothesis is supported by the observation that mutations that enlarge the active site cavity in proximity to the nucleoside 5-position endow dCK with the ability to phosphorylate thymidine. However, in conflict with this hypothesis was our discovery that the cytidine analogue 5-methyldeoxycytidine (5-Me-dC), an isostere of thymidine, can indeed be phosphorylated by wild-type (WT) dCK. To reconcile this seemingly contradicting observation, and to better understand the determinants preventing thymidine phosphorylation by WT dCK, we solved the crystal structure of dCK in complex with 5-Me-dC. The structure reveals the active site adjustments required to accommodate the methyl group at the 5-position. Combination of kinetic, mutagenesis, and structural data suggested that it is in fact residue Asp133 of dCK that is most responsible for discriminating against the thymine base. dCK variants in which Asp133 is replaced by an alanine and Arg104 by select hydrophobic residues attain significantly improved activity with 5-substituted deoxycytidine and thymidine analogues. Importantly, the ability of the designer enzymes to activate 5-substitued pyrimidines makes it possible to utilize such nucleoside analogues in suicide gene therapy or protein therapy applications that target cancer cells.


Assuntos
Desoxicitidina Quinase/química , Desoxicitidina/análogos & derivados , Desoxicitidina/metabolismo , Timidina/metabolismo , Substituição de Aminoácidos , Cristalografia por Raios X , Desoxicitidina Quinase/genética , Desoxicitidina Quinase/metabolismo , Humanos , Cinética , Fosforilação , Especificidade por Substrato , Timidina/análogos & derivados
12.
ACS Appl Bio Mater ; 3(5): 2986-2996, 2020 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-35025345

RESUMO

Composite bioceramic and hydrogel-based containers harboring alkaline phosphatase are generated through encapsulation of this enzyme by its immobilization into CaCO3-based bioceramic materials in combination with a hydrogel assembly technique and subsequent gelification. A refined way of synthesis and modification allows preparing the enzyme delivery system with functionalized protection layers. The particles are characterized by electron microscopy, Fourier transform infrared (FTIR) spectroscopy, and enzyme activity measurements. Loading efficiency and loading capacity are investigated depending on particle size, time of enzyme loading, and various container compositions and enzyme concentrations. Our results reveal that the size of particles influences their morphology and this, in turn, affects the activity of the encapsulated enzymes. Various functionalizations of the surfaces, including protection by the hydrogel layer, formation of hollow silver alginate, or calcium alginate encapsulation, decrease the enzymatic activity. The presence of a good therapeutic effect on osteoblastic cells coupled with a relatively high loading capacity, biocompatibility, and ease of fabrication suggests that the developed carriers are promising candidates for efficient drug delivery, especially in the field of bone reconstruction.

13.
Macromol Biosci ; 20(7): e2000081, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32484303

RESUMO

Encapsulation of enzymes allows to preserve their biological activities in various environmental conditions, such as exposure to elevated temperature or to proteases. This is particularly relevant for in vivo applications, where proteases represent a severe obstacle to maintaining the activity of enzymes. Polyelectrolyte multilayer capsules are suitable for enzyme encapsulation, where CaCO3 particles and temperature-dependent capsule formation are the best templates and the most adequate method, respectively. In this work, these two areas are combined and, ALP (alkaline phosphatase), which is a robust and therapeutically relevant enzyme, is encapsulated into thermally shrunk polyelectrolyte multilayer (PDADMAC/PSS)4 capsules templated on calcium carbonate particles (original average diameter: ≈3.5 µm). The activity of the encapsulated enzyme and the optimal temperature range for encapsulation are investigated. The enzymatic activity is almost four times higher upon encapsulation when the temperature range for encapsulation is situated just above the glass transition temperature (40 °C), while its optimal conditions are dictated, on the one hand, by the enzyme activity (better at lower temperatures) and, on the other hand, by the size and mechanical properties of capsules (better at higher temperatures).


Assuntos
Fosfatase Alcalina/metabolismo , Carbonato de Cálcio/química , Polieletrólitos/química , Temperatura , Cápsulas , Microscopia de Força Atômica , Tamanho da Partícula , Polietilenos/química , Compostos de Amônio Quaternário/química
14.
Biochemistry ; 48(6): 1256-63, 2009 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-19159229

RESUMO

Salvage of nucleosides in the cytosol of human cells is carried out by deoxycytidine kinase (dCK) and thymidine kinase 1 (TK1). Whereas TK1 is only responsible for thymidine phosphorylation, dCK is capable of converting dC, dA, and dG into their monophosphate forms. Using structural data on dCK, we predicted that select mutations at the active site would, in addition to making the enzyme faster, expand the catalytic repertoire of dCK to include thymidine. Specifically, we hypothesized that steric repulsion between the methyl group of the thymine base and Arg104 is the main factor preventing the phosphorylation of thymidine by wild-type dCK. Here we present kinetic data on several dCK variants where Arg104 has been replaced by select residues, all performed in combination with the mutation of Asp133 to an alanine. We show that several hydrophobic residues at position 104 endow dCK with thymidine kinase activity. Depending on the exact nature of the mutations, the enzyme's substrate preference is modified. The R104M-D133A double mutant is a pyrimidine-specific enzyme due to large K(m) values with purines. The crystal structure of the double mutant R104M-D133A in complex with the L-form of thymidine supplies a structural explanation for the ability of this variant to phosphorylate thymidine and thymidine analogs. The replacement of Arg104 by a smaller residue allows L-dT to bind deeper into the active site, making space for the C5-methyl group of the thymine base. The unique catalytic properties of several of the mutants make them good candidates for suicide-gene/protein-therapy applications.


Assuntos
Biocatálise , Desoxicitidina Quinase/metabolismo , Timidina Quinase/metabolismo , Difosfato de Adenosina , Cristalografia por Raios X , Citarabina/metabolismo , Desoxiadenosinas/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Desoxicitidina/metabolismo , Desoxiguanosina/metabolismo , Humanos , Cinética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação/genética , Estrutura Secundária de Proteína , Eletricidade Estática , Especificidade por Substrato , Timidina/metabolismo , Gencitabina
15.
Mol Microbiol ; 68(6): 1583-94, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18433446

RESUMO

Adenylate kinase (Adk1p) is a pivotal enzyme in both energetic and adenylic nucleotide metabolisms. In this paper, using a transcriptomic analysis, we show that the lack of Adk1p strongly induced expression of the PHO and ADE genes involved in phosphate utilization and AMP de novo biosynthesis respectively. Isolation and characterization of adk1 point mutants affecting PHO5 expression revealed that all these mutations also severely affected Adk1p catalytic activity, as well as PHO84 and ADE1 transcription. Furthermore, overexpression of distantly related enzymes such as human adenylate kinase or yeast UMP kinase was sufficient to restore regulation. These results demonstrate that adenylate kinase catalytic activity is critical for proper regulation of the PHO and ADE pathways. We also establish that adk1 deletion and purine limitation have similar effects on both adenylic nucleotide pool and PHO84 or ADE17 expression. Finally, we show that, in the adk1 mutant, upregulation of ADE1 depends on synthesis of the previously described effector(s) (S)AICAR ((N-succinyl)-5-aminoimidazol-4-carboxamide ribotide), while upregulation of PHO84 necessitates the Spl2p positive regulator. This work reveals that adenylic nucleotide availability is a key signal used by yeast to co-ordinate phosphate utilization and purine synthesis.


Assuntos
Nucleotídeos de Adenina/metabolismo , Adenilato Quinase/metabolismo , Regulação Fúngica da Expressão Gênica , Isoenzimas/metabolismo , Redes e Vias Metabólicas , Fosfatos/metabolismo , Purinas/metabolismo , Leveduras/enzimologia , Adenilato Quinase/genética , Proteínas Fúngicas , Perfilação da Expressão Gênica , Humanos , Inosina/metabolismo , Isoenzimas/genética , Mutação Puntual , Deleção de Sequência , Leveduras/genética , Leveduras/metabolismo
16.
Nucleic Acids Res ; 35(1): 186-92, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17158155

RESUMO

L-nucleoside analogs represent an important class of small molecules for treating both viral infections and cancers. These pro-drugs achieve pharmacological activity only after enzyme-catalyzed conversion to their tri-phosphorylated forms. Herein, we report the crystal structures of human deoxycytidine kinase (dCK) in complex with the L-nucleosides (-)-beta-2',3'-dideoxy-3'-thiacytidine (3TC)--an approved anti-human immunodeficiency virus (HIV) agent--and troxacitabine (TRO)--an experimental anti-neoplastic agent. The first step in activating these agents is catalyzed by dCK. Our studies reveal how dCK, which normally catalyzes phosphorylation of the natural D-nucleosides, can efficiently phosphorylate substrates with non-physiologic chirality. The capability of dCK to phosphorylate both D- and L-nucleosides and nucleoside analogs derives from structural properties of both the enzyme and the substrates themselves. First, the nucleoside-binding site tolerates substrates with different chiral configurations by maintaining virtually all of the protein-ligand interactions responsible for productive substrate positioning. Second, the pseudo-symmetry of nucleosides and nucleoside analogs in combination with their conformational flexibility allows the L- and D-enantiomeric forms to adopt similar shapes when bound to the enzyme. This is the first analysis of the structural basis for activation of L-nucleoside analogs, providing further impetus for discovery and clinical development of new agents in this molecular class.


Assuntos
Fármacos Anti-HIV/química , Antineoplásicos/química , Citosina/análogos & derivados , Desoxicitidina Quinase/química , Dioxolanos/química , Lamivudina/química , Modelos Moleculares , Cristalografia por Raios X , Citosina/química , Humanos , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Estereoisomerismo , Especificidade por Substrato
17.
Structure ; 15(12): 1555-66, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18073106

RESUMO

The human cytosolic thymidine kinase (TK) and structurally related TKs in prokaryotes play a crucial role in the synthesis and regulation of the cellular thymidine triphosphate pool. We report the crystal structures of the TK homotetramer from Thermotoga maritima in four different states: its apo-form, a binary complex with thymidine, as well as the ternary structures with the two substrates (thymidine/AppNHp) and the reaction products (TMP/ADP). In combination with fluorescence spectroscopy and mutagenesis experiments, our results demonstrate that ATP binding is linked to a substantial reorganization of the enzyme quaternary structure, leading to a transition from a closed, inactive conformation to an open, catalytic state. We hypothesize that these structural changes are relevant to enzyme function in situ as part of the catalytic cycle and serve an important role in regulating enzyme activity by amplifying the effects of feedback inhibitor binding.


Assuntos
Timidina Quinase/metabolismo , Trifosfato de Adenosina/metabolismo , Dissulfetos/química , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ligação Proteica , Estrutura Quaternária de Proteína , Espectrometria de Fluorescência , Timidina Quinase/química , Difração de Raios X
18.
Biochim Biophys Acta Biomembr ; 1861(11): 183026, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31465764

RESUMO

We investigated melting transitions in native biological membranes containing their membrane proteins. The membranes originated from E. coli, B. subtilis, lung surfactant and nerve tissue from the spinal cord of several mammals. For some preparations, we studied the pressure, pH and ionic strength dependence of the transition. For porcine spine, we compared the transition of the native membrane to that of the extracted lipids. All preparations displayed melting transitions of 10-20° below physiological or growth temperature, independent of the organism of origin and the respective cell type. We found that the position of the transitions in E. coli membranes depends on the growth temperature. We discuss these findings in the context of the thermodynamic theory of membrane fluctuations close to transition that predicts largely altered elastic constants, an increase in fluctuation lifetime and in membrane permeability. We also discuss how to distinguish lipid melting from protein unfolding transitions. Since the feature of a transition slightly below physiological temperature is conserved even when growth conditions change, we conclude that the transitions are likely to be of major biological importance for the survival and the function of the cell.


Assuntos
Membrana Celular/química , Membrana Celular/metabolismo , Temperatura de Transição , Animais , Bactérias , Fenômenos Fisiológicos Bacterianos , Membrana Celular/fisiologia , Lipídeos/análise , Mamíferos/fisiologia , Concentração Osmolar , Transição de Fase , Temperatura , Termodinâmica
19.
FEBS Lett ; 582(5): 720-4, 2008 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-18258203

RESUMO

Intracellular phosphorylation of dCK on Ser-74 results in increased nucleoside kinase activity. We mimicked this phosphorylation by a Ser-74-Glu mutation in bacterially produced dCK and investigated kinetic parameters using various nucleoside substrates. The S74E mutation increases the k(cat) values 11-fold for dC, and 3-fold for the anti-cancer analogues dFdC and AraC. In contrast, the rate is decreased for the purine substrates. In HEK293 cells, we found that by comparing transiently transfected dCK(S74E)-GFP and wild-type dCK-GFP, mimicking the phosphorylation of Ser-74 has no effect on cellular localisation. We note that phosphorylation may represent a mechanism to enhance the catalytic activity of the relatively slow dCK enzyme.


Assuntos
Desoxicitidina Quinase/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/metabolismo , Mimetismo Molecular , Fosfosserina/metabolismo , Transporte Ativo do Núcleo Celular , Catálise , Linhagem Celular , Núcleo Celular/enzimologia , Desoxicitidina Quinase/química , Humanos , Espaço Intracelular/enzimologia , Cinética , Proteínas Mutantes/metabolismo , Fosforilação , Estrutura Secundária de Proteína , Transporte Proteico , Especificidade por Substrato
20.
J Mol Biol ; 367(2): 488-500, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17276460

RESUMO

Bifunctional human PAPS synthetase (PAPSS) catalyzes, in a two-step process, the formation of the activated sulfate carrier 3'-phosphoadenosine 5'-phosphosulfate (PAPS). The first reaction involves the formation of the 5'-adenosine phosphosulfate (APS) intermediate from ATP and inorganic sulfate. APS is then further phosphorylated on its 3'-hydroxyl group by an additional ATP molecule to generate PAPS. The former reaction is catalyzed by the ATP-sulfurylase domain and the latter by the APS-kinase domain. Here, we report the structure of the APS-kinase domain of PAPSS isoform 1 (PAPSS1) representing the Michaelis complex with the products ADP-Mg and PAPS. This structure provides a rare glimpse of the active conformation of an enzyme catalyzing phosphoryl transfer without resorting to substrate analogs, inactivating mutations, or catalytically non-competent conditions. Our structure shows the interactions involved in the binding of the magnesium ion and PAPS, thereby revealing residues critical for catalysis. The essential magnesium ion is observed bridging the phosphate groups of the products. This function of the metal ion is made possible by the DGDN-loop changing its conformation from that previously reported, and identifies these loop residues unambiguously as a Walker B motif. Furthermore, the second aspartate residue of this motif is the likely candidate for initiating nucleophilic attack on the ATP gamma-phosphate group by abstracting the proton from the 3'-hydroxyl group of the substrate APS. We report the structure of the APS-kinase domain of human PAPSS1 in complex with two APS molecules, demonstrating the ability of the ATP/ADP-binding site to bind APS. Both structures reveal extended N termini that approach the active site of the neighboring monomer. Together, these results significantly increase our understandings of how catalysis is achieved by APS-kinase.


Assuntos
Adenosina Fosfossulfato/metabolismo , Magnésio/metabolismo , Modelos Moleculares , Complexos Multienzimáticos/química , Sulfato Adenililtransferase/química , Sequência de Aminoácidos , Sítios de Ligação , Cátions Bivalentes , Ativação Enzimática , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Dados de Sequência Molecular , Complexos Multienzimáticos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Sulfato Adenililtransferase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA