Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Inherit Metab Dis ; 46(6): 1170-1185, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37540500

RESUMEN

CAD is a large, 2225 amino acid multienzymatic protein required for de novo pyrimidine biosynthesis. Pathological CAD variants cause a developmental and epileptic encephalopathy which is highly responsive to uridine supplements. CAD deficiency is difficult to diagnose because symptoms are nonspecific, there is no biomarker, and the protein has over 1000 known variants. To improve diagnosis, we assessed the pathogenicity of 20 unreported missense CAD variants using a growth complementation assay that identified 11 pathogenic variants in seven affected individuals; they would benefit from uridine treatment. We also tested nine variants previously reported as pathogenic and confirmed the damaging effect of seven. However, we reclassified two variants as likely benign based on our assay, which is consistent with their long-term follow-up with uridine. We found that several computational methods are unreliable predictors of pathogenic CAD variants, so we extended the functional assay results by studying the impact of pathogenic variants at the protein level. We focused on CAD's dihydroorotase (DHO) domain because it accumulates the largest density of damaging missense changes. The atomic-resolution structures of eight DHO pathogenic variants, combined with functional and molecular dynamics analyses, provided a comprehensive structural and functional understanding of the activity, stability, and oligomerization of CAD's DHO domain. Combining our functional and protein structural analysis can help refine clinical diagnostic workflow for CAD variants in the genomics era.


Asunto(s)
Dihidroorotasa , Proteínas , Humanos , Dihidroorotasa/química , Dihidroorotasa/genética , Dihidroorotasa/metabolismo , Mutación Missense , Uridina
2.
Biomolecules ; 13(1)2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36671534

RESUMEN

Dihydroorotase (DHOase) is the third enzyme in the pathway used for the biosynthesis of pyrimidine nucleotides. In mammals, DHOase is active in a trifunctional enzyme, CAD, which also carries out the activities of carbamoyl phosphate synthetase and aspartate transcarbamoylase. Prior to this study, it was unknown whether the FDA-approved clinical drug 5-fluorouracil (5-FU), which is used as an anticancer therapy, could bind to the DHOase domain of human CAD (huDHOase). Here, we identified huDHOase as a new 5-FU binding protein, thereby extending the 5-FU interactome to this human enzyme. In order to investigate where 5-FU binds to huDHOase, we solved the complexed crystal structure at 1.97 Å (PDB ID 8GVZ). The structure of huDHOase complexed with malate was also determined for the sake of comparison (PDB ID 8GW0). These two nonsubstrate ligands were bound at the active site of huDHOase. It was previously established that the substrate N-carbamoyl-L-aspartate is either bound to or moves away from the active site, but it is the loop that is extended towards (loop-in mode) or moved away (loop-out mode) from the active site. DHOase also binds to nonsubstrate ligands via the loop-out mode. In contrast to the Escherichia coli DHOase model, our complexed structures revealed that huDHOase binds to either 5-FU or malate via the loop-in mode. We further characterized the binding of 5-FU to huDHOase using site-directed mutagenesis and the fluorescence quenching method. Considering the loop-in mode, the dynamic loop in huDHOase should be a suitable drug-targeting site for further designing inhibitors and clinical chemotherapies to suppress pyrimidine biosynthesis in cancer cell lines.


Asunto(s)
Antineoplásicos , Dihidroorotasa , Animales , Humanos , Dihidroorotasa/química , Dihidroorotasa/metabolismo , Malatos , Ligandos , Fluorouracilo/farmacología , Antineoplásicos/farmacología , Mamíferos/metabolismo
3.
Molecules ; 28(2)2023 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-36677714

RESUMEN

CAD is a 1.5 MDa hexameric protein with four enzymatic domains responsible for initiating de novo biosynthesis of pyrimidines nucleotides: glutaminase, carbamoyl phosphate synthetase, aspartate transcarbamoylase (ATC), and dihydroorotase. Despite its central metabolic role and implication in cancer and other diseases, our understanding of CAD is poor, and structural characterization has been frustrated by its large size and sensitivity to proteolytic cleavage. Recently, we succeeded in isolating intact CAD-like particles from the fungus Chaetomium thermophilum with high yield and purity, but their study by cryo-electron microscopy is hampered by the dissociation of the complex during sample grid preparation. Here we devised a specific crosslinking strategy to enhance the stability of this mega-enzyme. Based on the structure of the isolated C. thermophilum ATC domain, we inserted by site-directed mutagenesis two cysteines at specific locations that favored the formation of disulfide bridges and covalent oligomers. We further proved that this covalent linkage increases the stability of the ATC domain without damaging the structure or enzymatic activity. Thus, we propose that this cysteine crosslinking is a suitable strategy to strengthen the contacts between subunits in the CAD particle and facilitate its structural characterization.


Asunto(s)
Aspartato Carbamoiltransferasa , Ácido Aspártico , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/química , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/metabolismo , Microscopía por Crioelectrón , Proteínas , Dihidroorotasa/química , Aspartato Carbamoiltransferasa/genética , Aspartato Carbamoiltransferasa/química , Aspartato Carbamoiltransferasa/metabolismo
4.
Proteins ; 91(1): 91-98, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35978488

RESUMEN

In this paper, we report the structural analysis of dihydroorotase (DHOase) from the hyperthermophilic and barophilic archaeon Methanococcus jannaschii. DHOase catalyzes the reversible cyclization of N-carbamoyl-l-aspartate to l-dihydroorotate in the third step of de novo pyrimidine biosynthesis. DHOases form a very diverse family of enzymes and have been classified into types and subtypes with structural similarities and differences among them. This is the first archaeal DHOase studied by x-ray diffraction. Its structure and comparison with known representatives of the other subtypes help define the structural features of the archaeal subtype. The M. jannaschii DHOase is found here to have traits from all subtypes. Contrary to expectations, it has a carboxylated lysine bridging the two Zn ions in the active site, and a long catalytic loop. It is a monomeric protein with a large ß sandwich domain adjacent to the TIM barrel. Loop 5 is similar to bacterial type III and the C-terminal extension is long.


Asunto(s)
Dihidroorotasa , Methanocaldococcus , Dihidroorotasa/química , Dihidroorotasa/metabolismo , Methanocaldococcus/metabolismo , Dominio Catalítico , Catálisis , Ácido Aspártico
5.
Molecules ; 26(23)2021 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-34885830

RESUMEN

Dihydroorotase (DHOase), a dimetalloenzyme containing a carbamylated lysine within the active site, is a member of the cyclic amidohydrolase family, which also includes allantoinase (ALLase), dihydropyrimidinase (DHPase), hydantoinase, and imidase. Unlike most known cyclic amidohydrolases, which are tetrameric, DHOase exists as a monomer or dimer. Here, we report and analyze two crystal structures of the eukaryotic Saccharomyces cerevisiae DHOase (ScDHOase) complexed with malate. The structures of different DHOases were also compared. An asymmetric unit of these crystals contained four crystallographically independent ScDHOase monomers. ScDHOase shares structural similarity with Escherichia coli DHOase (EcDHOase). Unlike EcDHOase, ScDHOase can form tetramers, both in the crystalline state and in solution. In addition, the subunit-interacting residues of ScDHOase for dimerization and tetramerization are significantly different from those of other DHOases. The tetramerization pattern of ScDHOase is also different from those of DHPase and ALLase. Based on sequence analysis and structural evidence, we identify two unique helices (α6 and α10) and a loop (loop 7) for tetramerization, and discuss why the residues for tetramerization in ScDHOase are not necessarily conserved among DHOases.


Asunto(s)
Dihidroorotasa/química , Dihidroorotasa/metabolismo , Multimerización de Proteína , Saccharomyces cerevisiae/enzimología , Amidohidrolasas/química , Amidohidrolasas/metabolismo , Secuencia de Aminoácidos , Biocatálisis , Cristalografía por Rayos X , Estabilidad de Enzimas , Humanos , Enlace de Hidrógeno , Lisina/metabolismo , Malatos/metabolismo , Modelos Moleculares , Saccharomyces cerevisiae/genética , Soluciones , Temperatura
6.
Int J Mol Sci ; 22(18)2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34576147

RESUMEN

Drug-resistant Staphylococcus aureus is an imminent threat to public health, increasing the importance of drug discovery utilizing unexplored bacterial pathways and enzyme targets. De novo pyrimidine biosynthesis is a specialized, highly conserved pathway implicated in both the survival and virulence of several clinically relevant pathogens. Class I dihydroorotase (DHOase) is a separate and distinct enzyme present in gram positive bacteria (i.e., S. aureus, B. anthracis) that converts carbamoyl-aspartate (Ca-asp) to dihydroorotate (DHO)-an integral step in the de novo pyrimidine biosynthesis pathway. This study sets forth a high-throughput screening (HTS) of 3000 fragment compounds by a colorimetry-based enzymatic assay as a primary screen, identifying small molecule inhibitors of S. aureus DHOase (SaDHOase), followed by hit validation with a direct binding analysis using surface plasmon resonance (SPR). Competition SPR studies of six hit compounds and eight additional analogs with the substrate Ca-asp determined the best compound to be a competitive inhibitor with a KD value of 11 µM, which is 10-fold tighter than Ca-asp. Preliminary structure-activity relationship (SAR) provides the foundation for further structure-based antimicrobial inhibitor design against S. aureus.


Asunto(s)
Dihidroorotasa/antagonistas & inhibidores , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/farmacología , Staphylococcus aureus/enzimología , Dominio Catalítico , Dihidroorotasa/química , Dihidroorotasa/aislamiento & purificación , Dihidroorotasa/metabolismo , Inhibidores Enzimáticos/química , Simulación del Acoplamiento Molecular , Bibliotecas de Moléculas Pequeñas/química , Staphylococcus aureus/efectos de los fármacos , Relación Estructura-Actividad
7.
Protein Sci ; 30(10): 1995-2008, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34288185

RESUMEN

CAD is a 1.5 MDa particle formed by hexameric association of a 250 kDa protein divided into different enzymatic domains, each catalyzing one of the initial reactions for de novo biosynthesis of pyrimidine nucleotides: glutaminase-dependent Carbamoyl phosphate synthetase, Aspartate transcarbamoylase, and Dihydroorotase. The pathway for de novo pyrimidine synthesis is essential for cell proliferation and is conserved in all living organisms, but the covalent linkage of the first enzymatic activities into a multienzymatic CAD particle is unique to animals. In other organisms, these enzymatic activities are encoded as monofunctional proteins for which there is abundant structural and biochemical information. However, the knowledge about CAD is scarce and fragmented. Understanding CAD requires not only to determine the three-dimensional structures and define the catalytic and regulatory mechanisms of the different enzymatic domains, but also to comprehend how these domains entangle and work in a coordinated and regulated manner. This review summarizes significant progress over the past 10 years toward the characterization of CAD's architecture, function, regulatory mechanisms, and cellular compartmentalization, as well as the recent finding of a new and rare neurometabolic disorder caused by defects in CAD activities.


Asunto(s)
Aspartato Carbamoiltransferasa , Encefalopatías Metabólicas/enzimología , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante) , Dihidroorotasa , Animales , Aspartato Carbamoiltransferasa/química , Aspartato Carbamoiltransferasa/metabolismo , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/química , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/metabolismo , Dihidroorotasa/química , Dihidroorotasa/metabolismo , Humanos , Dominios Proteicos
8.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202294

RESUMEN

Dihydroorotase (DHOase) is the third enzyme in the de novo biosynthesis pathway for pyrimidine nucleotides, and an attractive target for potential anticancer chemotherapy. By screening plant extracts and performing GC-MS analysis, we identified and characterized that the potent anticancer drug plumbagin (PLU), isolated from the carnivorous plant Nepenthes miranda, was a competitive inhibitor of DHOase. We also solved the complexed crystal structure of yeast DHOase with PLU (PDB entry 7CA1), to determine the binding interactions and investigate the binding modes. Mutational and structural analyses indicated the binding of PLU to DHOase through loop-in mode, and this dynamic loop may serve as a drug target. PLU exhibited cytotoxicity on the survival, migration, and proliferation of 4T1 cells and induced apoptosis. These results provide structural insights that may facilitate the development of new inhibitors targeting DHOase, for further clinical anticancer chemotherapies.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Productos Biológicos/farmacología , Vías Biosintéticas/efectos de los fármacos , Dihidroorotasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Naftoquinonas/farmacología , Pirimidinas/biosíntesis , Antineoplásicos Fitogénicos/química , Sitios de Unión , Productos Biológicos/química , Dominio Catalítico , Dihidroorotasa/química , Dihidroorotasa/genética , Inhibidores Enzimáticos/química , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Mutación , Naftoquinonas/química , Unión Proteica , Relación Estructura-Actividad
9.
Biochem Biophys Res Commun ; 551: 33-37, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33714757

RESUMEN

Dihydroorotase (DHOase) is the third enzyme in the de novo biosynthesis pathway of pyrimidine nucleotides and considered an attractive target for potential antimalarial, anticancer, and antipathogen chemotherapy. Whether the FDA-approved clinical drug 5-fluorouracil (5-FU) that is used to target the enzyme thymidylate synthase for anticancer therapy can also bind to DHOase remains unknown. Here, we report the crystal structures of DHOase from Saccharomyces cerevisiae (ScDHOase) complexed with malate, 5-FU, and 5-aminouracil (5-AU). ScDHOase shares structural similarity with Escherichia coli DHOase. We also characterized the binding of 5-FU and 5-AU to ScDHOase by using the fluorescence quenching method. These complexed structures revealed that residues Arg18, Asn43, Thr106, and Ala275 of ScDHOase were involved in the 5-FU (PDB entry 6L0B) and 5-AU binding (PDB entry 6L0F). Overall, these results provide structural insights that may facilitate the development of new inhibitors targeting DHOase and constitute the 5-FU and 5-AU interactomes for further clinical chemotherapies.


Asunto(s)
Antineoplásicos/química , Dihidroorotasa/química , Fluorouracilo/química , Saccharomyces cerevisiae/enzimología , Uracilo/análogos & derivados , Antineoplásicos/farmacología , Sitios de Unión , Cristalización , Cristalografía por Rayos X , Dihidroorotasa/metabolismo , Escherichia coli/enzimología , Fluorouracilo/farmacología , Malatos/química , Modelos Moleculares , Unión Proteica , Uracilo/química , Uracilo/farmacología
10.
Cells ; 9(5)2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32370067

RESUMEN

Ebola virus (EBOV) is a zoonotic pathogen causing severe hemorrhagic fevers in humans and non-human primates with high case fatality rates. In recent years, the number and extent of outbreaks has increased, highlighting the importance of better understanding the molecular aspects of EBOV infection and host cell interactions to control this virus more efficiently. Many viruses, including EBOV, have been shown to recruit host proteins for different viral processes. Based on a genome-wide siRNA screen, we recently identified the cellular host factor carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD) as being involved in EBOV RNA synthesis. However, mechanistic details of how this host factor plays a role in the EBOV life cycle remain elusive. In this study, we analyzed the functional and molecular interactions between EBOV and CAD. To this end, we used siRNA knockdowns in combination with various reverse genetics-based life cycle modelling systems and additionally performed co-immunoprecipitation and co-immunofluorescence assays to investigate the influence of CAD on individual aspects of the EBOV life cycle and to characterize the interactions of CAD with viral proteins. Following this approach, we could demonstrate that CAD directly interacts with the EBOV nucleoprotein NP, and that NP is sufficient to recruit CAD into inclusion bodies dependent on the glutaminase (GLN) domain of CAD. Further, siRNA knockdown experiments indicated that CAD is important for both viral genome replication and transcription, while substrate rescue experiments showed that the function of CAD in pyrimidine synthesis is indeed required for those processes. Together, this suggests that NP recruits CAD into inclusion bodies via its GLN domain in order to provide pyrimidines for EBOV genome replication and transcription. These results define a novel mechanism by which EBOV hijacks host cell pathways in order to facilitate genome replication and transcription and provide a further basis for the development of host-directed broad-spectrum antivirals.


Asunto(s)
Aspartato Carbamoiltransferasa/metabolismo , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/metabolismo , Dihidroorotasa/metabolismo , Ebolavirus/fisiología , Genoma Viral , Cuerpos de Inclusión Viral/metabolismo , Nucleoproteínas/metabolismo , Transcripción Genética , Proteínas Virales/metabolismo , Replicación Viral , Animales , Aspartato Carbamoiltransferasa/química , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/química , Línea Celular , Dihidroorotasa/química , Ebolavirus/genética , Técnicas de Silenciamiento del Gen , Humanos , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Pirimidinas/farmacología , ARN/metabolismo
11.
PLoS One ; 15(3): e0229494, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32126100

RESUMEN

Pseudomonas aeruginosa is a virulent pathogen that has become more threatening with the emergence of multidrug resistance. The aspartate transcarbamoylase (ATCase) of this organism is a dodecamer comprised of six 37 kDa catalytic chains and six 45 kDa chains homologous to dihydroorotase (pDHO). The pDHO chain is inactive but is necessary for ATCase activity. A stoichiometric mixture of the subunits associates into a dodecamer with full ATCase activity. Unlike other known ATCases, the P. aeruginosa catalytic chain does not spontaneously assemble into a trimer. Chemical-crosslinking and size-exclusion chromatography showed that P. aeruginosa ATCase is monomeric which accounts for its lack of catalytic activity since the active site is a composite comprised of residues from adjacent monomers in the trimer. Circular dichroism spectroscopy indicated that the ATCase chain adopts a structure that contains secondary structure elements although neither the ATCase nor the pDHO subunits are very stable as determined by a thermal shift assay. Formation of the complex increases the melting temperature by about 30°C. The ATCase is strongly inhibited by all nucleotide di- and triphosphates and exhibits extreme cooperativity. Previous studies suggested that the regulatory site is located in an 11-residue extension of the amino end of the catalytic chain. However, deletion of the extensions did not affect catalytic activity, nucleotide inhibition or the assembly of the dodecamer. Nucleotides destabilized the dodecamer which probably accounts for the inhibition and apparent cooperativity of the substrate saturation curves. Contrary to previous interpretations, these results suggest that P. aeruginosa ATCase is not allosterically regulated by nucleotides.


Asunto(s)
Aspartato Carbamoiltransferasa/química , Aspartato Carbamoiltransferasa/metabolismo , Dihidroorotasa/química , Dihidroorotasa/metabolismo , Pseudomonas aeruginosa/enzimología , Secuencias de Aminoácidos , Aspartato Carbamoiltransferasa/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biocatálisis , Dominio Catalítico , Dicroismo Circular , Dihidroorotasa/genética , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/genética , Termodinámica
12.
Int J Biol Macromol ; 136: 1176-1187, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31207330

RESUMEN

The de novo pyrimidine biosynthesis pathway is essential for the proliferation of many pathogens. One of the pathway enzymes, dihydroorotase (DHO), catalyzes the reversible interconversion of N-carbamoyl-l-aspartate to 4,5-dihydroorotate. The substantial difference between bacterial and mammalian DHOs makes it a promising drug target for disrupting bacterial growth and thus an important candidate to evaluate as a response to antimicrobial resistance on a molecular level. Here, we present two novel three-dimensional structures of DHOs from Yersinia pestis (YpDHO), the plague-causing pathogen, and Vibrio cholerae (VcDHO), the causative agent of cholera. The evaluations of these two structures led to an analysis of all available DHO structures and their classification into known DHO types. Comparison of all the DHO active sites containing ligands that are listed in DrugBank was facilitated by a new interactive, structure-comparison and presentation platform. In addition, we examined the genetic context of characterized DHOs, which revealed characteristic patterns for different types of DHOs. We also generated a homology model for DHO from Plasmodium falciparum.


Asunto(s)
Dihidroorotasa/química , Dihidroorotasa/metabolismo , Pirimidinas/biosíntesis , Vibrio cholerae/enzimología , Yersinia pestis/enzimología , Secuencia de Aminoácidos , Dominio Catalítico , Dihidroorotasa/genética , Genómica , Malatos/metabolismo , Modelos Moleculares , Homología de Secuencia de Aminoácido , Zinc/metabolismo
13.
FEBS J ; 286(6): 1204-1213, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30657257

RESUMEN

Dihydroorotase (DHOase) is involved in the de novo synthesis of pyrimidine in virtually all organisms, and it is usually associated with two other enzymes found in this biosynthetic pathway, carbamylphosphate synthetase and/or aspartate transcarbamylase (ATCase). In the hyperthermophilic bacterium Aquifex aeolicus, ATCase and DHOase are noncovalently associated. Upon dissociation, ATCase keeps its activity entirely while DHOase is totally inactivated. It was previously shown that high pressure fully restores the activity of this isolated DHOase. On the basis of kinetic studies, site-directed mutagenesis and the use of peptides mimicking loop A, a loop that appears to block access to the active site, was proposed that this pressure-induced reactivation was due to the decrease in the volume of the system, -ΔV, resulting from the disruption of known ionic interactions between the loop and the main part of the protein. In this study, this interpretation is more precisely demonstrated by the determination of the crystallographic structure of isolated DHOase under pressure. In addition to the loop displacements, pressure induces a discrete rearrangement of the catalytic site aspartate 305, an effect that might additionally contribute to the reactivation of this enzyme.


Asunto(s)
Ácido Aspártico/metabolismo , Bacterias/enzimología , Dihidroorotasa/química , Dihidroorotasa/metabolismo , Zinc/metabolismo , Aquifex , Ácido Aspártico/química , Ácido Aspártico/genética , Dominio Catalítico , Cristalografía , Dihidroorotasa/genética , Mutagénesis Sitio-Dirigida , Mutación , Presión , Conformación Proteica
14.
Subcell Biochem ; 93: 505-538, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31939163

RESUMEN

CAD is a 1.5 MDa particle formed by hexameric association of a 250 kDa protein that carries the enzymatic activities for the first three steps in the de novo biosynthesis of pyrimidine nucleotides: glutamine-dependent Carbamoyl phosphate synthetase, Aspartate transcarbamoylase and Dihydroorotase. This metabolic pathway is essential for cell growth and proliferation and is conserved in all living organisms. However, the fusion of the first three enzymatic activities of the pathway into a single multienzymatic protein only occurs in animals. In prokaryotes, by contrast, these activities are encoded as distinct monofunctional enzymes that function independently or by forming more or less transient complexes. Whereas the structural information about these enzymes in bacteria is abundant, the large size and instability of CAD has only allowed a fragmented characterization of its structure. Here we retrace some of the most significant efforts to decipher the architecture of CAD and to understand its catalytic and regulatory mechanisms.


Asunto(s)
Aspartato Carbamoiltransferasa/metabolismo , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/metabolismo , Dihidroorotasa/metabolismo , Complejos Multienzimáticos/química , Complejos Multienzimáticos/metabolismo , Pirimidinas/biosíntesis , Animales , Aspartato Carbamoiltransferasa/química , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/química , Dihidroorotasa/química
15.
J Biol Chem ; 293(49): 18903-18913, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30315107

RESUMEN

The dihydroorotase (DHOase) domain of the multifunctional protein carbamoyl-phosphate synthetase 2, aspartate transcarbamoylase, and dihydroorotase (CAD) catalyzes the third step in the de novo biosynthesis of pyrimidine nucleotides in animals. The crystal structure of the DHOase domain of human CAD (huDHOase) revealed that, despite evolutionary divergence, its active site components are highly conserved with those in bacterial DHOases, encoded as monofunctional enzymes. An important element for catalysis, conserved from Escherichia coli to humans, is a flexible loop that closes as a lid over the active site. Here, we combined mutagenic, structural, biochemical, and molecular dynamics analyses to characterize the function of the flexible loop in the activity of CAD's DHOase domain. A huDHOase chimera bearing the E. coli DHOase flexible loop was inactive, suggesting the presence of distinctive elements in the flexible loop of huDHOase that cannot be replaced by the bacterial sequence. We pinpointed Phe-1563, a residue absolutely conserved at the tip of the flexible loop in CAD's DHOase domain, as a critical element for the conformational equilibrium between the two catalytic states of the protein. Substitutions of Phe-1563 with Ala, Leu, or Thr prevented the closure of the flexible loop and inactivated the protein, whereas substitution with Tyr enhanced the interactions of the loop in the closed position and reduced fluctuations and the reaction rate. Our results confirm the importance of the flexible loop in CAD's DHOase domain and explain the key role of Phe-1563 in configuring the active site and in promoting substrate strain and catalysis.


Asunto(s)
Aspartato Carbamoiltransferasa/química , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/química , Dihidroorotasa/química , Aspartato Carbamoiltransferasa/genética , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/genética , Catálisis , Dominio Catalítico , Dihidroorotasa/genética , Humanos , Simulación de Dinámica Molecular , Mutagénesis , Mutación , Fenilalanina/química , Conformación Proteica , Dominios Proteicos
16.
Biochem Biophys Res Commun ; 505(2): 439-444, 2018 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-30268498

RESUMEN

Dihydropyrimidinase (DHPase) is a member of the cyclic amidohydrolase family, which also includes allantoinase, dihydroorotase (DHOase), hydantoinase, and imidase. Almost all of these zinc metalloenzymes possess a binuclear metal center in which two metal ions are bridged by a post-translational carbamylated Lys. Crystal structure of Tetraodon nigroviridis DHPase reveals that one zinc ion is sufficient to stabilize Lys carbamylation. In this study, we found that one metal coordination was not sufficient to fix CO2 to the Lys in bacterial DHPase. We prepared and characterized mono-Zn DHPase from Pseudomonas aeruginosa (PaDHPase), and the catalytic activity of mono-Zn PaDHPase was not detected. The crystal structure of mono-Zn PaDHPase determined at 2.23 Šresolution (PDB entry 6AJD) revealed that Lys150 was no longer carbamylated. This finding indicated the decarbamylation of the Lys during the metal chelating process. To confirm the state of Lys carbamylation in mono-Zn PaDHPase in solution, mass spectrometric (MS) analysis was carried out. The MS result was in agreement with the theoretical value for uncarbamylated PaDHPase. Crystal structure of the human DHOase domain (huDHOase) K1556A mutant was also determined (PDB entry 5YNZ), and the structure revealed that the active site of huDHOase K1556A mutant contained one metal ion. Like mono-Zn PaDHPase, oxygen ligands of the carbamylated Lys were not required for Znα binding. Considering the collective data from X-ray crystal structure and MS analysis, mono-Zn PaDHPase in both crystalline state and solution was not carbamylated. In addition, structural evidences indicated that post-translational carbamylated Lys was not required for Znα binding in PaDHPase and in huDHOase.


Asunto(s)
Amidohidrolasas/química , Dihidroorotasa/química , Lisina/metabolismo , Carbamilación de Proteína , Proteínas Bacterianas/química , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Espectrometría de Masas , Proteínas Mutantes/química , Unión Proteica , Procesamiento Proteico-Postraduccional , Pseudomonas aeruginosa/enzimología , Zinc/metabolismo
17.
Protein J ; 36(4): 361-373, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28660315

RESUMEN

The gene that codes for the putative dihydroorotase in the hyperthermophilic archaeon Methanococcus jannaschii was subcloned in pET-21a and expressed in Escherichia coli. A purification protocol was devised. The purity of the protein was evaluated by SDS-PAGE and the protein was confirmed by sequencing using LC-MS. The calculated molecular mass is 48104 Da. SEC-LS suggested that the protein is a monomer in solution. ICP-MS showed that there are two Zn ions per monomer. Kinetic analysis of the recombinant protein gave hyperbolic kinetics with Vmax = 12.2 µmol/min/mg and Km = 0.14 mM at 25 °C. Furthermore the activity of the protein increased with temperature consistent with the hyperthermophilic nature of the organism. A homology model was constructed using the mesophilic Bacillus anthracis protein as the template. Residues known to be critical for Zn and substrate binding were conserved. The activity of the enzyme at 85 and 90 °C was found to be relatively constant over 160 min and this correlates with the temperature of optimal growth of the organism of 85 °C. The amino acid sequences and structures of the two proteins were compared and this gave insight into some of the factors that may confer thermostability-more Lys and Ile, fewer Ala, Thr, Gln and Gly residues, and shorter N- and C-termini. Additional and better insight into the thermostabilization strategies adopted by this enzyme will be provided when its crystal structure is determined.


Asunto(s)
Proteínas Arqueales/química , Dihidroorotasa/química , Methanocaldococcus/química , Zinc/química , Secuencia de Aminoácidos , Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Bacillus anthracis/química , Bacillus anthracis/enzimología , Sitios de Unión , Clonación Molecular , Secuencia Conservada , Dihidroorotasa/genética , Dihidroorotasa/metabolismo , Estabilidad de Enzimas , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Cinética , Methanocaldococcus/enzimología , Peso Molecular , Sistemas de Lectura Abierta , Plásmidos/química , Plásmidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Relación Estructura-Actividad , Especificidad por Sustrato , Termodinámica , Transformación Bacteriana , Zinc/metabolismo
18.
Structure ; 25(6): 912-923.e5, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28552578

RESUMEN

CAD, the multifunctional protein initiating and controlling de novo biosynthesis of pyrimidines in animals, self-assembles into ∼1.5 MDa hexamers. The structures of the dihydroorotase (DHO) and aspartate transcarbamoylase (ATC) domains of human CAD have been previously determined, but we lack information on how these domains associate and interact with the rest of CAD forming a multienzymatic unit. Here, we prove that a construct covering human DHO and ATC oligomerizes as a dimer of trimers and that this arrangement is conserved in CAD-like from fungi, which holds an inactive DHO-like domain. The crystal structures of the ATC trimer and DHO-like dimer from the fungus Chaetomium thermophilum confirm the similarity with the human CAD homologs. These results demonstrate that, despite being inactive, the fungal DHO-like domain has a conserved structural function. We propose a model that sets the DHO and ATC complex as the central element in the architecture of CAD.


Asunto(s)
Aspartato Carbamoiltransferasa/química , Aspartato Carbamoiltransferasa/metabolismo , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/química , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/metabolismo , Dihidroorotasa/química , Dihidroorotasa/metabolismo , Aspartato Carbamoiltransferasa/genética , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/genética , Carbamoil Fosfato/química , Carbamoil Fosfato/metabolismo , Chaetomium/enzimología , Cristalografía por Rayos X , Dihidroorotasa/genética , Humanos , Microscopía Electrónica , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Dominios Proteicos , Multimerización de Proteína , Pirimidinas/biosíntesis
19.
Curr Mol Med ; 17(1): 60-69, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28231751

RESUMEN

BACKGROUND: STIM/ORAI-mediated store-operated Ca2+ entry (SOCE) mediates a myriad of Ca2+-dependent cellular activities in mammals. Genetic defects in STIM1/ORAI1 lead to devastating severe combined immunodeficiency; whereas gain-offunction mutations in STIM1/ORAI1 are intimately associated with tubular aggregate myopathy. At molecular level, a decrease in the Ca2+ concentrations within the lumen of endoplasmic reticulum (ER) initiates multimerization of the STIM1 luminal domain to switch on the STIM1 cytoplasmic domain to engage and gate ORAI channels, thereby leading to the ultimate Ca2+ influx from the extracellular space into the cytosol. Despite tremendous progress made in dissecting functional STIM1-ORAI1 coupling, the activation mechanism of SOCE remains to be fully characterized. OBJECTIVE AND METHODS: Building upon a robust fluorescence resonance energy transfer assay designed to monitor STIM1 intramolecular autoinhibition, we aimed to systematically dissect the molecular determinants required for the activation and oligomerization of STIM1. RESULTS: Here we showed that truncation of the STIM1 luminal domain predisposes STIM1 to adopt a more active conformation. Replacement of the single transmembrane (TM) domain of STIM1 by a more rigid dimerized TM domain of glycophorin A abolished STIM1 activation. But this adverse effect could be partially reversed by disrupting the TM dimerization interface. Moreover, our study revealed regions that are important for the optimal assembly of hetero-oligomers composed of full-length STIM1 with its minimal STIM1-ORAI activating region, SOAR. CONCLUSIONS: Our study clarifies the roles of major STIM1 functional domains in maintaining a quiescent configuration of STIM1 to prevent preactivation of SOCE.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Secuencia de Aminoácidos , Aspartato Carbamoiltransferasa/química , Aspartato Carbamoiltransferasa/metabolismo , Señalización del Calcio , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/química , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/metabolismo , Línea Celular , Dihidroorotasa/química , Dihidroorotasa/metabolismo , Humanos , Activación del Canal Iónico , Microscopía Confocal , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Transducción de Señal , Molécula de Interacción Estromal 1/agonistas , Molécula de Interacción Estromal 1/química , Relación Estructura-Actividad
20.
J Biol Chem ; 292(2): 629-637, 2017 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-27746403

RESUMEN

Elevated hydrostatic pressure was used to probe conformational changes of Aquifex aeolicus dihydroorotase (DHO), which catalyzes the third step in de novo pyrimidine biosynthesis. The isolated protein, a 45-kDa monomer, lacks catalytic activity but becomes active upon formation of a dodecameric complex with aspartate transcarbamoylase (ATC). X-ray crystallographic studies of the isolated DHO and of the complex showed that association induces several major conformational changes in the DHO structure. In the isolated DHO, a flexible loop occludes the active site blocking the access of substrates. The loop is mostly disordered but is tethered to the active site region by several electrostatic and hydrogen bonds. This loop becomes ordered and is displaced from the active site upon formation of DHO-ATC complex. The application of pressure to the complex causes its time-dependent dissociation and the loss of both DHO and ATC activities. Pressure induced irreversible dissociation of the obligate ATC trimer, and as a consequence the DHO is also inactivated. However, moderate hydrostatic pressure applied to the isolated DHO subunit mimics the complex formation and reversibly activates the isolated subunit in the absence of ATC, suggesting that the loop has been displaced from the active site. This effect of pressure is explained by the negative volume change associated with the disruption of ionic interactions and exposure of ionized amino acids to the solvent (electrostriction). The interpretation that the loop is relocated by pressure was validated by site-directed mutagenesis and by inhibition by small peptides that mimic the loop residues.


Asunto(s)
Aspartato Carbamoiltransferasa/metabolismo , Bacterias/enzimología , Proteínas Bacterianas/metabolismo , Dihidroorotasa/metabolismo , Multimerización de Proteína/fisiología , Aspartato Carbamoiltransferasa/química , Aspartato Carbamoiltransferasa/genética , Bacterias/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Dominio Catalítico/fisiología , Dihidroorotasa/química , Dihidroorotasa/genética , Activación Enzimática/fisiología , Presión Hidrostática
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...