Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Mol Microbiol ; 122(2): 201-212, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38922722

RESUMEN

An arsenate reductase (Car1) from the Bacteroidetes species Rufibacter tibetensis 1351T was isolated from the Tibetan Plateau. The strain exhibits resistance to arsenite [As(III)] and arsenate [As(V)] and reduces As(V) to As(III). Here we shed light on the mechanism of enzymatic reduction by Car1. AlphaFold2 structure prediction, active site energy minimization, and steady-state kinetics of wild-type and mutant enzymes give insight into the catalytic mechanism. Car1 is structurally related to calcineurin-like metallophosphoesterases (MPPs). It functions as a binuclear metal hydrolase with limited phosphatase activity, particularly relying on the divalent metal Ni2+. As an As(V) reductase, it displays metal promiscuity and is coupled to the thioredoxin redox cycle, requiring the participation of two cysteine residues, Cys74 and Cys76. These findings suggest that Car1 evolved from a common ancestor of extant phosphatases by incorporating a redox function into an existing MPP catalytic site. Its proposed mechanism of arsenate reduction involves Cys74 initiating a nucleophilic attack on arsenate, leading to the formation of a covalent intermediate. Next, a nucleophilic attack of Cys76 leads to the release of As(III) and the formation of a surface-exposed Cys74-Cys76 disulfide, ready for reduction by thioredoxin.


Asunto(s)
Arseniato Reductasas , Bacteroidetes , Dominio Catalítico , Oxidación-Reducción , Arseniato Reductasas/metabolismo , Arseniato Reductasas/genética , Arseniato Reductasas/química , Bacteroidetes/enzimología , Bacteroidetes/genética , Arseniatos/metabolismo , Cinética , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/química , Catálisis , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Arsenitos/metabolismo
2.
Mol Cell ; 68(3): 552-565.e8, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29056322

RESUMEN

mTOR complex I (mTORC1) is a central growth regulator that senses amino acids through a pathway that converges on the Rag GTPases, an obligate heterodimer of two related GTPases. Despite their central role in amino acid sensing, it is unknown why the Rag GTPases are heterodimeric and whether their subunits communicate with each other. Here, we find that the binding of guanosine triphosphate (GTP) to one subunit inhibits the binding and induces the hydrolysis of GTP by the other. This intersubunit communication pushes the Rag GTPases into either of two stable configurations, which represent active "on" or "off" states that interconvert via transient intermediates. Subunit coupling confers on the mTORC1 pathway its capacity to respond rapidly to the amino acid level. Thus, the dynamic response of mTORC1 requires intersubunit communication by the Rag GTPases, providing a rationale for why they exist as a dimer and revealing a distinct mode of control for a GTP-binding protein.


Asunto(s)
Aminoácidos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Sitios de Unión , Estabilidad de Enzimas , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Hidrólisis , Cinética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Modelos Moleculares , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/genética , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Subunidades de Proteína , Transducción de Señal , Relación Estructura-Actividad , Transfección
3.
Int J Mol Sci ; 25(18)2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39337255

RESUMEN

Nucleoside diphosphate kinases (NDPKs) are encoded by nme genes and exist in various isoforms. Based on interactions with other proteins, they are involved in signal transduction, development and pathological processes such as tumorigenesis, metastasis and heart failure. In this study, we report a 1.25 Å resolution structure of human homohexameric NDPK-C bound to ADP and describe the yet unknown complexes formed with GDP, UDP and cAMP, all obtained at a high resolution via X-ray crystallography. Each nucleotide represents a distinct group of mono- or diphosphate purine or pyrimidine bases. We analyzed different NDPK-C nucleotide complexes in the presence and absence of Mg2+ and explain how this ion plays an essential role in NDPKs' phosphotransferase activity. By analyzing a nucleotide-depleted NDPK-C structure, we detected conformational changes upon substrate binding and identify flexible regions in the substrate binding site. A comparison of NDPK-C with other human isoforms revealed a strong similarity in the overall composition with regard to the 3D structure, but significant differences in the charge and hydrophobicity of the isoforms' surfaces. This may play a role in isoform-specific NDPK interactions with ligands and/or important complex partners like other NDPK isoforms, as well as monomeric and heterotrimeric G proteins. Considering the recently discovered role of NDPK-C in different pathologies, these high-resolution structures thus might provide a basis for interaction studies with other proteins or small ligands, like activators or inhibitors.


Asunto(s)
Nucleósido Difosfato Quinasas NM23 , Humanos , Adenosina Difosfato/metabolismo , Adenosina Difosfato/química , Sitios de Unión , Cristalografía por Rayos X , AMP Cíclico/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Difosfato/química , Magnesio/metabolismo , Magnesio/química , Modelos Moleculares , Nucleósido Difosfato Quinasas NM23/metabolismo , Nucleósido Difosfato Quinasas NM23/química , Nucleósido Difosfato Quinasas NM23/genética , Nucleósido-Difosfato Quinasa/química , Nucleósido-Difosfato Quinasa/metabolismo , Nucleósido-Difosfato Quinasa/genética , Nucleótidos/metabolismo , Nucleótidos/química , Unión Proteica , Conformación Proteica , Especificidad por Sustrato , Uridina Difosfato/metabolismo , Uridina Difosfato/química
4.
J Biol Chem ; 297(3): 101043, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34358565

RESUMEN

A large number of protein sequences are registered in public databases such as PubMed. Functionally uncharacterized enzymes are included in these databases, some of which likely have potential for industrial applications. However, assignment of the enzymes remained difficult tasks for now. In this study, we assigned a total of 28 original sequences to uncharacterized enzymes in the FAD-dependent oxidase family expressed in some species of bacteria including Chryseobacterium, Flavobacterium, and Pedobactor. Progenitor sequence of the assigned 28 sequences was generated by ancestral sequence reconstruction, and the generated sequence exhibited L-lysine oxidase activity; thus, we named the enzyme AncLLysO. Crystal structures of ligand-free and ligand-bound forms of AncLLysO were determined, indicating that the enzyme recognizes L-Lys by hydrogen bond formation with R76 and E383. The binding of L-Lys to AncLLysO induced dynamic structural change at a plug loop formed by residues 251 to 254. Biochemical assays of AncLLysO variants revealed the functional importance of these substrate recognition residues and the plug loop. R76A and E383D variants were also observed to lose their activity, and the kcat/Km value of G251P and Y253A mutations were approximately 800- to 1800-fold lower than that of AncLLysO, despite the indirect interaction of the substrates with the mutated residues. Taken together, our data demonstrate that combinational approaches to sequence classification from database and ancestral sequence reconstruction may be effective not only to find new enzymes using databases of unknown sequences but also to elucidate their functions.


Asunto(s)
Aminoácido Oxidorreductasas/química , Aminoácido Oxidorreductasas/metabolismo , Bacterias/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Aminoácido Oxidorreductasas/genética , Bacterias/química , Bacterias/genética , Proteínas Bacterianas/genética , Sitios de Unión , Catálisis , Minería de Datos , Enlace de Hidrógeno , Cinética , Lisina/química , Lisina/metabolismo , Modelos Moleculares
5.
J Biol Chem ; 295(33): 11379-11387, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32571879

RESUMEN

Systemic amyloidosis caused by extracellular deposition of insoluble fibrils derived from the pathological aggregation of circulating proteins, such as transthyretin, is a severe and usually fatal condition. Elucidation of the molecular pathogenic mechanism of the disease and discovery of effective therapies still represents a challenging medical issue. The in vitro preparation of amyloid fibrils that exhibit structural and biochemical properties closely similar to those of natural fibrils is central to improving our understanding of the biophysical basis of amyloid formation in vivo and may offer an important tool for drug discovery. Here, we compared the morphology and thermodynamic stability of natural transthyretin fibrils with those of fibrils generated in vitro either using the common acidification procedure or primed by limited selective cleavage by plasmin. The free energies for fibril formation were -12.36, -8.10, and -10.61 kcal mol-1, respectively. The fibrils generated via plasmin cleavage were more stable than those prepared at low pH and were thermodynamically and morphologically similar to natural fibrils extracted from human amyloidotic tissue. Determination of thermodynamic stability is an important tool that is complementary to other methods of structural comparison between ex vivo fibrils and fibrils generated in vitro Our finding that fibrils created via an in vitro amyloidogenic pathway are structurally similar to ex vivo human amyloid fibrils does not necessarily establish that the fibrillogenic pathway is the same for both, but it narrows the current knowledge gap between in vitro models and in vivo pathophysiology.


Asunto(s)
Neuropatías Amiloides Familiares/patología , Amiloide/química , Prealbúmina/química , Amiloide/genética , Amiloide/ultraestructura , Neuropatías Amiloides Familiares/genética , Humanos , Mutación , Prealbúmina/genética , Agregado de Proteínas , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Estabilidad Proteica , Termodinámica
6.
Zhongguo Zhong Yao Za Zhi ; 45(24): 5884-5889, 2020 Dec.
Artículo en Zh | MEDLINE | ID: mdl-33496128

RESUMEN

Protoberberine alkaloids belong to the quaternary ammonium isoquinoline alkaloids, and are the main active ingredients in traditional Chinese herbal medicines, like Coptis chinensis. They have been widely used to treat such diseases as gastroenteritis, intestinal infections, and conjunctivitis. Studies have shown that structural modification of the protoberberine alkaloids could produce derivative compounds with new pharmacological effects and biological activities, but the transformation mechanism is not clear yet. This article mainly summarizes the researches on the biotransformation and structure modification of protoberberine alkaloids mainly based on berberine, so as to provide background basis and new ideas for studies relating to the mechanism of protoberberine alkaloids and the pharmacological activity and application of new compounds.


Asunto(s)
Alcaloides , Alcaloides de Berberina , Berberina , Coptis , Biotransformación
7.
J Biol Chem ; 293(37): 14192-14199, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30018138

RESUMEN

Systemic amyloidosis is a usually fatal disease caused by extracellular accumulation of abnormal protein fibers, amyloid fibrils, derived by misfolding and aggregation of soluble globular plasma protein precursors. Both WT and genetic variants of the normal plasma protein transthyretin (TTR) form amyloid, but neither the misfolding leading to fibrillogenesis nor the anatomical localization of TTR amyloid deposition are understood. We have previously shown that, under physiological conditions, trypsin cleaves human TTR in a mechano-enzymatic mechanism that generates abundant amyloid fibrils in vitro In sharp contrast, the widely used in vitro model of denaturation and aggregation of TTR by prolonged exposure to pH 4.0 yields almost no clearly defined amyloid fibrils. However, the exclusive duodenal location of trypsin means that this enzyme cannot contribute to systemic extracellular TTR amyloid deposition in vivo Here, we therefore conducted a bioinformatics search for systemically active tryptic proteases with appropriate tissue distribution, which unexpectedly identified plasmin as the leading candidate. We confirmed that plasmin, just as trypsin, selectively cleaves human TTR between residues 48 and 49 under physiological conditions in vitro Truncated and full-length protomers are then released from the native homotetramer and rapidly aggregate into abundant fibrils indistinguishable from ex vivo TTR amyloid. Our findings suggest that physiological fibrinolysis is likely to play a critical role in TTR amyloid formation in vivo Identification of this surprising intersection between two hitherto unrelated pathways opens new avenues for elucidating the mechanisms of TTR amyloidosis, for seeking susceptibility risk factors, and for therapeutic innovation.


Asunto(s)
Amiloidosis/metabolismo , Plasminógeno/metabolismo , Prealbúmina/metabolismo , Amiloide/metabolismo , Bases de Datos de Proteínas , Fibrinolisina/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Desnaturalización Proteica , Pliegue de Proteína , Proteolisis , Tripsina/metabolismo
8.
Biochem J ; 475(13): 2167-2177, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29875256

RESUMEN

Biosynthesis of the gibberellin A (GA) plant hormones evolved independently in plant-associated fungi and bacteria. While the relevant enzymes have distinct evolutionary origins, the pathways proceed via highly similar reactions. One particularly complex transformation involves combined demethylation and γ-lactone ring formation, catalyzed in bacteria by the cytochrome P450 CYP112 in three individual steps, which involves large structural changes in the transition from substrate to product, with further divergence in the recently demonstrated use of two separate mechanistic routes. Here, the substrate specificity of the isozyme from Erwinia tracheiphila, EtCYP112, was probed via UV-Vis spectral binding studies and activity assays with alternate substrates from the GA biosynthetic pathway. EtCYP112 tightly binds its native substrate GA12 and reaction intermediates GA15 and GA24, as well as the methylated derivatives of GA12 and GA15 It, however, only poorly binds methylated GA24, its GA9 final product and the C-20 carboxylate side product GA25 These distinct affinities are consistent with the known reactivity of EtCYP112. However, while it binds to the immediately preceding pathway metabolite GA12-aldehyde and even earlier oxygenated ent-kaurene precursors, EtCYP112 only reacts with GA12-aldehyde and not the earlier ent-kaurene-derived metabolites. Even with GA12-aldehyde conversion is limited to the first two steps, and the full combined demethylation and γ-lactone ring-forming transformation is not catalyzed. Thus, CYP112 has evolved specificity at the catalytic rather than substrate-binding level to enable its role in GA biosynthesis.


Asunto(s)
Proteínas Bacterianas , Sistema Enzimático del Citocromo P-450 , Erwinia/enzimología , Sesquiterpenos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/metabolismo , Sesquiterpenos/química , Sesquiterpenos/metabolismo , Especificidad por Sustrato
9.
Proc Natl Acad Sci U S A ; 113(9): E1190-9, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26884191

RESUMEN

O-glycosylation of Ser and Thr residues is an important process in all organisms, which is only poorly understood. Such modification is required for the export and function of adhesin proteins that mediate the attachment of pathogenic Gram-positive bacteria to host cells. Here, we have analyzed the mechanism by which the cytosolic O-glycosyltransferase GtfA/B of Streptococcus gordonii modifies the Ser/Thr-rich repeats of adhesin. The enzyme is a tetramer containing two molecules each of GtfA and GtfB. The two subunits have the same fold, but only GtfA contains an active site, whereas GtfB provides the primary binding site for adhesin. During a first phase of glycosylation, the conformation of GtfB is restrained by GtfA to bind substrate with unmodified Ser/Thr residues. In a slow second phase, GtfB recognizes residues that are already modified with N-acetylglucosamine, likely by converting into a relaxed conformation in which one interface with GtfA is broken. These results explain how the glycosyltransferase modifies a progressively changing substrate molecule.


Asunto(s)
Adhesión Bacteriana , Proteínas Bacterianas/biosíntesis , Citosol/enzimología , Glicosiltransferasas/metabolismo , Proteínas Bacterianas/química , Cristalografía por Rayos X , Dimerización , Glicosiltransferasas/química , Bacterias Grampositivas/metabolismo , Modelos Moleculares , Conformación Proteica
10.
BMC Biochem ; 17(1): 12, 2016 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-27974044

RESUMEN

BACKGROUND: Nucleoside triphosphate (NTP) hydrolysis is a key reaction in biology. It involves breaking two very stable bonds (one P-O bond and one O-H bond of water), in either a concurrent or a sequential way. Here, we systematically examine how protonation of the triphosphate affects the mechanism of hydrolysis. RESULTS: The hydrolysis reaction of methyl triphosphate in vacuum is computed with protons in various numbers and position on the three phosphate groups. Protonation is seen to have a strong catalytic effect, with the reaction mechanism depending highly on the protonation pattern. CONCLUSION: This dependence is apparently complicated, but is shown to obey a well-defined set of rules: Protonation of the α- and ß-phosphate groups favors a sequential hydrolysis mechanism, whereas γ-protonation favors a concurrent mechanism, the two effects competing with each other in cases of simultaneous protonation. The rate-limiting step is always the breakup of the water molecule while it attacks the γ-phosphorus, and its barrier is lowered by γ-protonation. This step has significantly lower barriers in the sequential reactions, because the dissociated γ-metaphosphate intermediate (PγO3-) is a much better target for water attack than the un-dissociated γ-phosphate (-PγO42-). The simple chemical logic behind these rules helps to better understand the catalytic strategy used by NTPase enzymes, as illustrated here for the catalytic pocket of myosin. A set of rules was determined that describes how protonating the phosphate groups affects the hydrolysis mechanism of methyl triphosphate: Protonation of the α- and/or ß- phosphate groups promotes a sequential mechanism in which P-O bond breaking precedes the breakup of the attacking water, whereas protonation of the γ-phosphate promotes a concurrent mechanism and lowers the rate-limiting barrier of water breakup. The role played by individual protein residues in the catalytic pocket of triphosphate hydrolysing enzymes can be assigned accordingly.


Asunto(s)
Ácido Anhídrido Hidrolasas/metabolismo , Polifosfatos/metabolismo , Ácido Anhídrido Hidrolasas/química , Adenosina Trifosfato/metabolismo , Biocatálisis , Dominio Catalítico , Hidrólisis , Protones , Termodinámica , Vacio
11.
Chem Phys Lett ; 666: 58-61, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28603294

RESUMEN

Ribonuclease A is the oldest model for studying enzymatic mechanisms, yet questions remain about proton transfer within the active site. Seminal work by Jackson et al. (Science, 1994) labeled Ribonuclease A with 4-fluorohistidine, concluding that active-site histidines act as general acids and bases. Calculations of 4-fluorohistidine indicate that the π-tautomer is predominant in all simulated environments (by ~17 kJ/mol), strongly suggesting that fluoro-labeled ribonuclease A functions with His119 in π-tautomer. The tautomeric form of His119 during proton transfer and tautomerism as a putative mechanistic step in wild-type RNase A remain open questions and should be considered in future mechanistic studies.

12.
Arch Toxicol ; 90(12): 3017-3027, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26838043

RESUMEN

Microsomal epoxide hydrolase (mEH) is a detoxifying enzyme for xenobiotic compounds. Enzymatic activity of mEH can be greatly increased by a point mutation, leading to an E404D amino acid exchange in its catalytic triad. Surprisingly, this variant is not found in any vertebrate species, despite the obvious advantage of accelerated detoxification. We hypothesized that this evolutionary avoidance is due to the fact that the mEH plays a dualistic role in detoxification and control of endogenous vascular signaling molecules. To test this, we generated mEH E404D mice and assessed them for detoxification capacity and vascular dynamics. In liver microsomes from these mice, turnover of the xenobiotic compound phenanthrene-9,10-oxide was four times faster compared to WT liver microsomes, confirming accelerated detoxification. mEH E404D animals also showed faster metabolization of a specific class of endogenous eicosanoids, arachidonic acid-derived epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acids (DHETs). Significantly higher DHETs/EETs ratios were found in mEH E404D liver, urine, plasma, brain and cerebral endothelial cells compared to WT controls, suggesting a broad impact of the mEH mutant on endogenous EETs metabolism. Because EETs are strong vasodilators in cerebral vasculature, hemodynamics were assessed in mEH E404D and WT cerebral cortex and hippocampus using cerebral blood volume (CBV)-based functional magnetic resonance imaging (fMRI). Basal CBV0 levels were similar between mEH E404D and control mice in both brain areas. But vascular reactivity and vasodilation in response to the vasodilatory drug acetazolamide were reduced in mEH E404D forebrain compared to WT controls by factor 3 and 2.6, respectively. These results demonstrate a critical role for mEH E404D in vasodynamics and suggest that deregulation of endogenous signaling pathways is the undesirable gain of function associated with the E404D variant.


Asunto(s)
Circulación Cerebrovascular , Trastornos Cerebrovasculares/metabolismo , Epóxido Hidrolasas/metabolismo , Microsomas Hepáticos/enzimología , Mutación Puntual , Xenobióticos/farmacocinética , Sustitución de Aminoácidos , Animales , Dominio Catalítico , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Circulación Cerebrovascular/efectos de los fármacos , Trastornos Cerebrovasculares/genética , Trastornos Cerebrovasculares/fisiopatología , Eicosanoides/sangre , Eicosanoides/metabolismo , Eicosanoides/orina , Epóxido Hidrolasas/química , Epóxido Hidrolasas/genética , Hipocampo/irrigación sanguínea , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Inactivación Metabólica , Ratones , Ratones Mutantes , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fenantrenos/metabolismo , Resistencia Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Xenobióticos/metabolismo
13.
J Biol Chem ; 289(31): 21242-51, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24914210

RESUMEN

Mycobacterium tuberculosis (Mtb) synthesizes polymethylated polysaccharides that form complexes with long chain fatty acids. These complexes, referred to as methylglucose lipopolysaccharides (MGLPs), regulate fatty acid biosynthesis in vivo, including biosynthesis of mycolic acids that are essential for building the cell wall. Glucosyl-3-phosphoglycerate phosphatase (GpgP, EC 5.4.2.1), encoded by Rv2419c gene, catalyzes the second step of the pathway for the biosynthesis of MGLPs. The molecular basis for this dephosphorylation is currently not understood. Here, we describe the crystal structures of apo-, vanadate-bound, and phosphate-bound MtbGpgP, depicting unliganded, reaction intermediate mimic, and product-bound views of MtbGpgP, respectively. The enzyme consists of a single domain made up of a central ß-sheet flanked by α-helices on either side. The active site is located in a positively charged cleft situated above the central ß-sheet. Unambiguous electron density for vanadate covalently bound to His(11), mimicking the phosphohistidine intermediate, was observed. The role of residues interacting with the ligands in catalysis was probed by site-directed mutagenesis. Arg(10), His(11), Asn(17), Gln(23), Arg(60), Glu(84), His(159), and Leu(209) are important for enzymatic activity. Comparison of the structures of MtbGpgP revealed conformational changes in a key loop region connecting ß1 with α1. This loop regulates access to the active site. MtbGpgP functions as dimer. L209E mutation resulted in monomeric GpgP, rendering the enzyme incapable of dephosphorylation. The structures of GpgP reported here are the first crystal structures for histidine-phosphatase-type GpgPs. These structures shed light on a key step in biosynthesis of MGLPs that could be targeted for development of anti-tuberculosis therapeutics.


Asunto(s)
Ácidos Glicéricos/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Dimerización , Modelos Moleculares , Sistemas de Lectura Abierta , Fosforilación
14.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 7): 1832-43, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25004961

RESUMEN

Lysobacter enzymogenes lysyl endoproteinase (LysC) is a trypsin-type serine protease with a high pH optimum that hydrolyses all Lys-Xaa peptide bonds. The high specificity of LysC renders it useful for biotechnological purposes. The K30R variant of a related lysyl endoproteinase from Achromobacter lyticus has favourable enzymatic properties that might be transferrable to LysC. To visualize structural differences in the substrate-binding sites, the crystal structures of wild-type and the K30R variant of LysC were determined. The mutation is located at a distance of 12 Šfrom the catalytic triad and subtly changes the surface properties of the substrate-binding site. The high pH optimum of LysC can be attributed to electrostatic effects of an aromatic Tyr/His stack on the catalytic aspartate and is a general feature of this enzyme subfamily. LysC crystals in complex with the covalent inhibitor N(α)-p-tosyl-lysyl chloromethylketone yielded data to 1.1 and 0.9 Šresolution, resulting in unprecedented precision of the active and substrate-binding sites for this enzyme subfamily. Error estimates on bond lengths and difference electron density indicate that instead of the expected oxyanion a hydroxyl group binds to the partially solvent-exposed oxyanion hole. Protonation of the alkoxide catalytic intermediate might be a recurring feature during serine protease catalysis.


Asunto(s)
Radical Hidroxilo/metabolismo , Lisina/metabolismo , Lysobacter/enzimología , Péptido Hidrolasas/metabolismo , Aniones , Sitios de Unión , Cristalografía por Rayos X , Péptido Hidrolasas/química
15.
FASEB J ; 27(6): 2431-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23457216

RESUMEN

Sulfs are extracellular sulfatases that have emerged recently as critical regulators of heparan sulfate (HS) activities through their ability to catalyze specific 6-O-desulfation of the polysaccharide. Consequently, Sulfs have been involved in many physiological and pathological processes, and notably for Sulf-2, in the development of cancers with poor prognosis. Despite growing interest, little is known about the structure and activity of these enzymes and the way they induce dynamic remodeling of HS 6-O-sulfation status. Here, we have combined an array of analytical approaches, including mass spectrometry, NMR, HS oligosaccharide sequencing, and FACS, to dissect HSulf-2 sulfatase activity, either on a purified octasaccharide used as a mimic of HS functional domains, or on intact cell-surface HS chains. In parallel, we have studied the functional consequences of HSulf-2 activity on fibroblast growth factor (FGF)-induced mitogenesis and found that the enzyme could differentially regulate FGF1 and FGF2 activities. Notably, these data supported the existence of precise 6-O-sulfation patterns for FGF activation and provided new insights into the saccharide structures involved. Altogether, our data bring to light an original processive enzymatic mechanism, by which HSulfs catalyze oriented alteration of HS 6-O-desulfation patterns and direct fine and differential regulation of HS functions.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Heparitina Sulfato/metabolismo , Sulfotransferasas/metabolismo , Catálisis , Línea Celular , Heparitina Sulfato/química , Humanos , Oligosacáridos/química , Oligosacáridos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Especificidad por Sustrato , Sulfatasas , Sulfotransferasas/química
16.
RNA Biol ; 11(12): 1540-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25616362

RESUMEN

Pseudouridine (Ψ) is the most abundant of >150 nucleoside modifications in RNA. Although Ψ was discovered as the first modified nucleoside more than half a century ago, neither the enzymatic mechanism of its formation, nor the function of this modification are fully elucidated. We present the consistent picture of Ψ synthases, their substrates and their substrate positions in model organisms of all domains of life as it has emerged to date and point out the challenges that remain concerning higher eukaryotes and the elucidation of the enzymatic mechanism.


Asunto(s)
Transferasas Intramoleculares/metabolismo , Seudouridina/metabolismo , Procesamiento Postranscripcional del ARN , ARN Guía de Kinetoplastida/metabolismo , ARN de Transferencia Aminoácido-Específico/metabolismo , Uridina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Transferasas Intramoleculares/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Conformación de Ácido Nucleico , ARN/genética , ARN/metabolismo , ARN Guía de Kinetoplastida/química , ARN Guía de Kinetoplastida/genética , ARN Mitocondrial , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , ARN de Transferencia Aminoácido-Específico/química , ARN de Transferencia Aminoácido-Específico/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Ribosomas/química , Ribosomas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
17.
ACS Appl Mater Interfaces ; 16(22): 28452-28460, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38775640

RESUMEN

The electrocatalytic nitrogen reduction reaction (NRR) presents an alternative method for the Haber-Bosch process, and single-atom catalysts (SACs) to achieve efficient NRR have attracted considerable attention in the past decades. However, whether SACs are more suitable for NRR compared to atomic-cluster catalysts (ACCs) remains to be studied. Herein, we have successfully synthesized both the Fe monomers (Fe1) and trimers (Fe3) on nitrogen-doped carbon catalysts. Both the experiments and DFT calculations indicate that compared to the end-on adsorption of N2 on Fe1 catalysts, N2 activation is enhanced via the side-on adsorption on Fe3 catalysts, and the reaction follows the enzymatic pathway with a reduced free energy barrier for NRR. As a result, the Fe3 catalysts achieved better NRR performance (NH3 yield rate of 27.89 µg h-1 mg-1cat. and Faradaic efficiency of 45.13%) than Fe1 catalysts (10.98 µg h-1 mg-1cat. and 20.98%). Therefore, our research presents guidance to prepare more efficient NRR catalysts.

18.
Front Chem ; 12: 1373312, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38456185

RESUMEN

L-Asparaginases, divided into three structural Classes, catalyze the hydrolysis of L-asparagine to L-aspartic acid and ammonia. The members of Class 3, ReAIV and ReAV, encoded in the genome of the nitrogen fixing Rhizobium etli, have the same fold, active site, and quaternary structure, despite low sequence identity. In the present work we examined the biochemical consequences of this difference. ReAIV is almost twice as efficient as ReAV in asparagine hydrolysis at 37°C, with the kinetic KM, kcat parameters (measured in optimal buffering agent) of 1.5 mM, 770 s-1 and 2.1 mM, 603 s-1, respectively. The activity of ReAIV has a temperature optimum at 45°C-55°C, whereas the activity of ReAV, after reaching its optimum at 37°C, decreases dramatically at 45°C. The activity of both isoforms is boosted by 32 or 56%, by low and optimal concentration of zinc, which is bound three times more strongly by ReAIV then by ReAV, as reflected by the KD values of 1.2 and 3.3 µM, respectively. We also demonstrate that perturbation of zinc binding by Lys→Ala point mutagenesis drastically decreases the enzyme activity but also changes the mode of response to zinc. We also examined the impact of different divalent cations on the activity, kinetics, and stability of both isoforms. It appeared that Ni2+, Cu2+, Hg2+, and Cd2+ have the potential to inhibit both isoforms in the following order (from the strongest to weakest inhibitors) Hg2+ > Cu2+ > Cd2+ > Ni2+. ReAIV is more sensitive to Cu2+ and Cd2+, while ReAV is more sensitive to Hg2+ and Ni2+, as revealed by IC50 values, melting scans, and influence on substrate specificity. Low concentration of Cd2+ improves substrate specificity of both isoforms, suggesting its role in substrate recognition. The same observation was made for Hg2+ in the case of ReAIV. The activity of the ReAV isoform is less sensitive to Cl- anions, as reflected by the IC50 value for NaCl, which is eightfold higher for ReAV relative to ReAIV. The uncovered complementary properties of the two isoforms help us better understand the inducibility of the ReAV enzyme.

19.
Antibiotics (Basel) ; 12(12)2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-38136683

RESUMEN

Alginates play an important role in the resistance of mucoid strains of Pseudomonas aeruginosa to antibiotics, as well as their persistence by escaping the immune defense system. GDP-mannose dehydrogenase (GMD) is the key enzyme in alginate biosynthesis by catalyzing the irreversible double oxidation of GDP-mannose to GDP-mannuronate. GDP-mannose dehydrogenase purified from mucoid strains exhibits strong negative cooperativity for its substrate, the GDP-mannose, with a KM of 13 µM for the site of strong affinity and 3 mM for this weak of a binding. The presence of a nucleotide strongly associated with the enzyme was detected, confirming the fact that the substrate oxidation reaction takes place in two distinct steps, with the substrate blocked on the enzyme in a half-oxidation state in the form of a hemiacetal. As the GMD polypeptide has only one site for substrate binding, our results tend to confirm the fact that the enzyme functions in a dimer form. The GDP-mannose dehydrogenase inhibition strategy that we developed a few years ago, based on the synthesis of substrate analogs, has shown its effectiveness. The addition of an alkynyl radical on carbon 6 of the mannose grafted to an amino-sulfonyl-guanosine allows, at a concentration of 0.5 mM, to inhibit GMD by 90%. As we had previously shown the effectiveness of these analogs on the sensitivity of mucoid strains of Pseudomonas aeruginosa to aminoglycosides, this revives the interest in the synthesis of new inhibitors of GDP-mannose dehydrogenase.

20.
Acta Crystallogr D Struct Biol ; 79(Pt 8): 775-791, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37494066

RESUMEN

The genome of Rhizobium etli, a nitrogen-fixing bacterial symbiont of legume plants, encodes two L-asparaginases, ReAIV and ReAV, that have no similarity to the well characterized enzymes of class 1 (bacterial type) and class 2 (plant type). It has been hypothesized that ReAIV and ReAV might belong to the same structural class 3 despite their low level of sequence identity. When the crystal structure of the inducible and thermolabile protein ReAV was solved, this hypothesis gained a stronger footing because the key residues of ReAV are also present in the sequence of the constitutive and thermostable ReAIV protein. High-resolution crystal structures of ReAIV now confirm that it is a class 3 L-asparaginase that is structurally similar to ReAV but with important differences. The most striking differences concern the peculiar hydration patterns of the two proteins, the presence of three internal cavities in ReAIV and the behavior of the zinc-binding site. ReAIV has a high pH optimum (9-11) and a substrate affinity of ∼1.3 mM at pH 9.0. These parameters are not suitable for the direct application of ReAIV as an antileukemic drug, although its thermal stability and lack of glutaminase activity would be of considerable advantage. The five crystal structures of ReAIV presented in this work allow a possible enzymatic scenario to be postulated in which the zinc ion coordinated in the active site is a dispensable element. The catalytic nucleophile seems to be Ser47, which is part of two Ser-Lys tandems in the active site. The structures of ReAIV presented here may provide a basis for future enzyme-engineering experiments to improve the kinetic parameters for medicinal applications.


Asunto(s)
Asparaginasa , Rhizobium etli , Asparaginasa/química , Rhizobium etli/química , Rhizobium etli/genética , Catálisis , Sitios de Unión , Plantas/metabolismo , Zinc
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda