Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 14(3): 4285-4296, 2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35020352

RESUMEN

Scalability, process control, and modularity are some of the advantages that make flow biocatalysis a key-enabling technology for green and sustainable chemistry. In this context, rigid porous solid membranes hold the promise to expand the toolbox of flow biocatalysis due to their chemical stability and inertness. Yttrium-stabilized zirconia (YSZ) fulfills these properties; however, it has been scarcely exploited as a carrier for enzymes. Here, we discovered an unprecedented interaction between YSZ materials and His-tagged enzymes that enables the fabrication of multifunctional biocatalytic membranes for bioredox cascades. X-ray photoelectron spectroscopy suggests that enzyme immobilization is driven by coordination interactions between the imidazole groups of His-tags and both Zr and Y atoms. As model enzymes, we coimmobilized in-flow a thermophilic hydroxybutyryl-CoA dehydrogenase (TtHBDH-His) and a formate dehydrogenase (His-CbFDH) for the continuous asymmetric reduction of ethyl acetoacetate with in situ redox cofactor recycling. Fluorescence confocal microscopy deciphered the spatial organization of the two coimmobilized enzymes, pointing out the importance of the coimmobilization sequence. Finally, the coimmobilized system succeeded in situ, recycling the redox cofactor, maintaining the specific productivity using only 0.05 mM NADH, and accumulating a total enzyme turnover number of 4000 in 24 h. This work presents YSZ materials as ready-to-use carriers for the site-directed enzyme in-flow immobilization and the application of the resulting heterogeneous biocatalysts for continuous biomanufacturing.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Materiales Biocompatibles/metabolismo , Formiato Deshidrogenasas/metabolismo , Itrio/metabolismo , Circonio/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/química , Materiales Biocompatibles/química , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Formiato Deshidrogenasas/química , Ensayo de Materiales , Itrio/química , Circonio/química
2.
Nat Struct Mol Biol ; 28(9): 713-723, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34489609

RESUMEN

Human mitochondrial transcripts contain messenger and ribosomal RNAs flanked by transfer RNAs (tRNAs), which are excised by mitochondrial RNase (mtRNase) P and Z to liberate all RNA species. In contrast to nuclear or bacterial RNase P, mtRNase P is not a ribozyme but comprises three protein subunits that carry out RNA cleavage and methylation by unknown mechanisms. Here, we present the cryo-EM structure of human mtRNase P bound to precursor tRNA, which reveals a unique mechanism of substrate recognition and processing. Subunits TRMT10C and SDR5C1 form a subcomplex that binds conserved mitochondrial tRNA elements, including the anticodon loop, and positions the tRNA for methylation. The endonuclease PRORP is recruited and activated through interactions with its PPR and nuclease domains to ensure precise pre-tRNA cleavage. The structure provides the molecular basis for the first step of RNA processing in human mitochondria.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , Metiltransferasas/química , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN , Ribonucleasa P/química , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Anticodón/química , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Microscopía por Crioelectrón , Humanos , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Mitocondrias/enzimología , Modelos Moleculares , Mutación Missense , Conformación de Ácido Nucleico , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , ARN de Hongos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ribonucleasa P/metabolismo , Especificidad de la Especie , Relación Estructura-Actividad , Especificidad por Sustrato
3.
J Struct Biol ; 213(3): 107776, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34371166

RESUMEN

The Mycobacterium tuberculosis trifunctional enzyme (MtTFE) is an α2ß2 tetrameric enzyme. The α-chain harbors the 2E-enoyl-CoA hydratase (ECH) and 3S-hydroxyacyl-CoA dehydrogenase (HAD) activities and the ß-chain provides the 3-ketoacyl-CoA thiolase (KAT) activity. Enzyme kinetic data reported here show that medium and long chain enoyl-CoA molecules are preferred substrates for MtTFE. Modelling studies indicate how the linear medium and long acyl chains of these substrates can bind to each of the active sites. In addition, crystallographic binding studies have identified three new CoA binding sites which are different from the previously known CoA binding sites of the three TFE active sites. Structure comparisons provide new insights into the properties of ECH, HAD and KAT active sites of MtTFE. The interactions of the adenine moiety of CoA with loop-2 of the ECH active site cause a conformational change of this loop by which a competent ECH active site is formed. The NAD+ binding domain (domain C) of the HAD part of MtTFE has only a few interactions with the rest of the complex and adopts a range of open conformations, whereas the A-domain of the ECH part is rigidly fixed with respect to the HAD part. Two loops, the CB1-CA1 region and the catalytic CB4-CB5 loop, near the thiolase active site and the thiolase dimer interface, have high B-factors. Structure comparisons suggest that a competent and stable thiolase dimer is formed only when complexed with the α-chains, highlighting the importance of the assembly for the proper functioning of the complex.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas , Mycobacterium tuberculosis , 3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Enoil-CoA Hidratasa/química , Oxidación-Reducción , Especificidad por Sustrato
4.
Cell Death Dis ; 11(7): 563, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32703935

RESUMEN

17-beta-hydroxysteroid dehydrogenase 10 (HSD17B10) plays an important role in mitochondrial fatty acid metabolism and is also involved in mitochondrial tRNA maturation. HSD17B10 missense mutations cause HSD10 mitochondrial disease (HSD10MD). HSD17B10 with mutations identified from cases of HSD10MD show loss of function in dehydrogenase activity and mitochondrial tRNA maturation, resulting in mitochondrial dysfunction. It has also been implicated to play roles in the development of Alzheimer disease (AD) and tumorigenesis. Here, we found that HSD17B10 is a new substrate of NAD-dependent deacetylase Sirtuin 3 (SIRT3). HSD17B10 is acetylated at lysine residues K79, K99 and K105 by the acetyltransferase CBP, and the acetylation is reversed by SIRT3. HSD17B10 acetylation regulates its enzymatic activity and the formation of mitochondrial RNase P. Furthermore, HSD17B10 acetylation regulates the intracellular functions, affecting cell growth and cell resistance in response to stresses. Our results demonstrated that acetylation is an important regulation mechanism for HSD17B10 and may provide insight into interrupting the development of AD.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Estrés Oxidativo , Sirtuina 3/metabolismo , Estrés Fisiológico , 3-Hidroxiacil-CoA Deshidrogenasas/química , Acetilación , Empalme Alternativo/genética , Secuencia de Aminoácidos , Proliferación Celular , Células HCT116 , Células HEK293 , Humanos , Mitocondrias/metabolismo , Fragmentos de Péptidos/metabolismo , Unión Proteica , ARN de Transferencia/genética , Sialoglicoproteínas/metabolismo
5.
J Neurochem ; 155(3): 231-249, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32306391

RESUMEN

17ß-hydroxysteroid dehydrogenase (17ß-HSD10) is a multifunctional human enzyme with important roles both as a structural component and also as a catalyst of many metabolic pathways. This mitochondrial enzyme has important functions in the metabolism, development and aging of the neural system, where it is involved in the homeostasis of neurosteroids, especially in regard to estradiol, changes in which make it an essential part of neurodegenerative pathology. These roles therefore, indicate that 17ß-HSD10 may be a possible druggable target for neurodegenerative diseases including Alzheimer's disease (AD), and in hormone-dependent cancer. The objective of this review was to provide a summary about physiological functions and pathological roles of 17ß-HSD10 and the modulators of its activity.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/genética , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Estado de Salud , Mitocondrias/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/química , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Humanos , Mitocondrias/genética , Mutación/genética , Neoplasias/genética , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Estructura Secundaria de Proteína
6.
Int J Mol Sci ; 21(6)2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32192199

RESUMEN

Human 17ß-hydroxysteroid dehydrogenase type 10 is a multifunctional protein involved in many enzymatic and structural processes within mitochondria. This enzyme was suggested to be involved in several neurological diseases, e.g., mental retardation, Parkinson's disease, or Alzheimer's disease, in which it was shown to interact with the amyloid-beta peptide. We prepared approximately 60 new compounds based on a benzothiazolyl scaffold and evaluated their inhibitory ability and mechanism of action. The most potent inhibitors contained 3-chloro and 4-hydroxy substitution on the phenyl ring moiety, a small substituent at position 6 on the benzothiazole moiety, and the two moieties were connected via a urea linker (4at, 4bb, and 4bg). These compounds exhibited IC50 values of 1-2 µM and showed an uncompetitive mechanism of action with respect to the substrate, acetoacetyl-CoA. These uncompetitive benzothiazolyl inhibitors of 17ß-hydroxysteroid dehydrogenase type 10 are promising compounds for potential drugs for neurodegenerative diseases that warrant further research and development.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/antagonistas & inhibidores , Benzotiazoles/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Urea/química , Urea/farmacología , 3-Hidroxiacil-CoA Deshidrogenasas/química , Enfermedad de Alzheimer/tratamiento farmacológico , Activación Enzimática , Humanos , Cinética , Estructura Molecular , Proteínas Recombinantes , Relación Estructura-Actividad
7.
Acta Crystallogr D Struct Biol ; 75(Pt 1): 101-108, 2019 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-30644849

RESUMEN

The intracellular pathogen Mycobacterium tuberculosis is the causative agent of tuberculosis, which is a leading cause of mortality worldwide. The survival of M. tuberculosis in host macrophages through long-lasting periods of persistence depends, in part, on breaking down host cell lipids as a carbon source. The critical role of fatty-acid catabolism in this organism is underscored by the extensive redundancy of the genes implicated in ß-oxidation (∼100 genes). In a previous study, the enzymology of the M. tuberculosis L-3-hydroxyacyl-CoA dehydrogenase FadB2 was characterized. Here, the crystal structure of this enzyme in a ligand-free form is reported at 2.1 Šresolution. FadB2 crystallized as a dimer with three unique dimer copies per asymmetric unit. The structure of the monomer reveals a dual Rossmann-fold motif in the N-terminal domain, while the helical C-terminal domain mediates dimer formation. Comparison with the CoA- and NAD+-bound human orthologue mitochondrial hydroxyacyl-CoA dehydrogenase shows extensive conservation of the residues that mediate substrate and cofactor binding. Superposition with the multi-catalytic homologue M. tuberculosis FadB, which forms a trifunctional complex with the thiolase FadA, indicates that FadB has developed structural features that prevent its self-association as a dimer. Conversely, FadB2 is unable to substitute for FadB in the tetrameric FadA-FadB complex as it lacks the N-terminal hydratase domain of FadB. Instead, FadB2 may functionally (or physically) associate with the enoyl-CoA hydratase EchA8 and the thiolases FadA2, FadA3, FadA4 or FadA6 as suggested by interrogation of the STRING protein-network database.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , Mycobacterium tuberculosis/enzimología , Cristalografía por Rayos X , Enoil-CoA Hidratasa/metabolismo , Humanos , Oxidación-Reducción , Unión Proteica , Multimerización de Proteína
8.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 11): 733-740, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30387779

RESUMEN

(S)-3-Hydroxybutyryl-CoA dehydrogenase (HBD) has been gaining increased attention recently as it is a key enzyme in the enantiomeric formation of (S)-3-hydroxybutyryl-CoA [(S)-3HB-CoA]. It converts acetoacetyl-CoA to (S)-3HB-CoA in the synthetic metabolic pathway. (S)-3HB-CoA is further modified to form (S)-3-hydroxybutyrate, which is a source of biodegradable polymers. During the course of a study to develop biodegradable polymers, attempts were made to determine the crystal structure of HBD from Clostridium acetobutylicum (CacHBD), and the crystal structures of both apo and NAD+-bound forms of CacHBD were determined. The crystals belonged to different space groups: P212121 and P21. However, both structures adopted a hexamer composed of three dimers in the asymmetric unit, and this oligomerization was additionally confirmed by gel-filtration column chromatography. Furthermore, to investigate the catalytic residues of CacHBD, the enzymatic activities of the wild type and of three single-amino-acid mutants were analyzed, in which the Ser, His and Asn residues that are conserved in the HBDs from C. acetobutylicum, C. butyricum and Ralstonia eutropha, as well as in the L-3-hydroxyacyl-CoA dehydrogenases from Homo sapiens and Escherichia coli, were substituted by alanines. The S117A and N188A mutants abolished the activity, while the H138A mutant showed a slightly lower Km value and a significantly lower kcat value than the wild type. Therefore, in combination with the crystal structures, it was shown that His138 is involved in catalysis and that Ser117 and Asn188 may be important for substrate recognition to place the keto group of the substrate in the correct position for reaction.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/genética , Sustitución de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Dominio Catalítico , Clostridium acetobutylicum , Cristalografía por Rayos X , Cinética , Modelos Moleculares , Mutación , NAD/química , NAD/metabolismo , Conformación Proteica , Multimerización de Proteína , Subunidades de Proteína/química
9.
J Biol Chem ; 293(33): 12862-12876, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29880640

RESUMEN

Mitochondrial tRNAs are transcribed as long polycistronic transcripts of precursor tRNAs and undergo posttranscriptional modifications such as endonucleolytic processing and methylation required for their correct structure and function. Among them, 5'-end processing and purine 9 N1-methylation of mitochondrial tRNA are catalyzed by two proteinaceous complexes with overlapping subunit composition. The Mg2+-dependent RNase P complex for 5'-end cleavage comprises the methyltransferase domain-containing protein tRNA methyltransferase 10C, mitochondrial RNase P subunit (TRMT10C/MRPP1), short-chain oxidoreductase hydroxysteroid 17ß-dehydrogenase 10 (HSD17B10/MRPP2), and metallonuclease KIAA0391/MRPP3. An MRPP1-MRPP2 subcomplex also catalyzes the formation of 1-methyladenosine/1-methylguanosine at position 9 using S-adenosyl-l-methionine as methyl donor. However, a lack of structural information has precluded insights into how these complexes methylate and process mitochondrial tRNA. Here, we used a combination of X-ray crystallography, interaction and activity assays, and small angle X-ray scattering (SAXS) to gain structural insight into the two tRNA modification complexes and their components. The MRPP1 N terminus is involved in tRNA binding and monomer-monomer self-interaction, whereas the C-terminal SPOUT fold contains key residues for S-adenosyl-l-methionine binding and N1-methylation. The entirety of MRPP1 interacts with MRPP2 to form the N1-methylation complex, whereas the MRPP1-MRPP2-MRPP3 RNase P complex only assembles in the presence of precursor tRNA. This study proposes low-resolution models of the MRPP1-MRPP2 and MRPP1-MRPP2-MRPP3 complexes that suggest the overall architecture, stoichiometry, and orientation of subunits and tRNA substrates.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , Metiltransferasas/química , Modelos Moleculares , Complejos Multienzimáticos/química , ARN Mitocondrial/química , ARN de Transferencia/química , Ribonucleasa P/química , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Cristalografía por Rayos X , Humanos , Metiltransferasas/metabolismo , Complejos Multienzimáticos/metabolismo , ARN Mitocondrial/metabolismo , ARN de Transferencia/metabolismo , Ribonucleasa P/metabolismo , Dispersión del Ángulo Pequeño
10.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3294-3302, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28888424

RESUMEN

MRPP2 (also known as HSD10/SDR5C1) is a multifunctional protein that harbours both catalytic and non-catalytic functions. The protein belongs to the short-chain dehydrogenase/reductases (SDR) family and is involved in the catabolism of isoleucine in vivo and steroid metabolism in vitro. MRPP2 also moonlights in a complex with the MRPP1 (also known as TRMT10C) protein for N1-methylation of purines at position 9 of mitochondrial tRNA, and in a complex with MRPP1 and MRPP3 (also known as PRORP) proteins for 5'-end processing of mitochondrial precursor tRNA. Inherited mutations in the HSD17B10 gene encoding MRPP2 protein lead to a childhood disorder characterised by progressive neurodegeneration, cardiomyopathy or both. Here we report two patients with novel missense mutations in the HSD17B10 gene (c.34G>C and c.526G>A), resulting in the p.V12L and p.V176M substitutions. Val12 and Val176 are highly conserved residues located at different regions of the MRPP2 structure. Recombinant mutant proteins were expressed and characterised biochemically to investigate their effects towards the functions of MRPP2 and associated complexes in vitro. Both mutant proteins showed significant reduction in the dehydrogenase, methyltransferase and tRNA processing activities compared to wildtype, associated with reduced stability for protein with p.V12L, whereas the protein carrying p.V176M showed impaired kinetics and complex formation. This study therefore identified two distinctive molecular mechanisms to explain the biochemical defects for the novel missense patient mutations.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/genética , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Mitocondrias/metabolismo , ARN de Transferencia/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/química , Femenino , Expresión Génica , Humanos , Lactante , Masculino , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Moleculares , Mutación Missense , Conformación Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ribonucleasa P/genética , Ribonucleasa P/metabolismo
11.
Chem Biol Drug Des ; 90(6): 1041-1055, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28660722

RESUMEN

Present work aimed to introduce non-peptidic ABAD loop D (LD ) hot spot mimetics as ABAD-Aß inhibitors. A full-length atomistic model of ABAD-Aß complex was built as a scaffold to launch the lead design and its topology later verified by cross-checking the computational mutagenesis results with that of in vitro data. Thereafter, the interactions of prime Aß-binding LD residues-Tyr101, Thr108, and Thr110-were translated into specific pharmacophore features and this hypothesis subsequently used as a virtual screen query. ELISA-based screening of 20 hits identified two promising lead candidates, VC15 and VC19 with an IC50 of 4.4 ± 0.3 and 9.6 ± 0.1 µm, respectively. They productively reversed Aß-induced mitochondrial dysfunctions such as mitochondrial membrane potential loss (JC-1 assay), toxicity (MTT assay), and ATP reduction (ATP assay) in addition to increased cell viabilities. This is the first reporting of LD hot spot-centric in silico scheme to discover novel compounds with promising ABAD-Aß inhibitory potential. These chemotypes are proposed for further structural optimization to derive novel Alzheimer's disease (AD) therapeutics.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Péptidos beta-Amiloides/toxicidad , Compuestos Heterocíclicos de 4 o más Anillos/metabolismo , Mitocondrias/efectos de los fármacos , 3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/genética , Adenosina Trifosfato/metabolismo , Enfermedad de Alzheimer , Animales , Sitios de Unión , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Semivida , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/toxicidad , Humanos , Concentración 50 Inhibidora , Ligandos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/metabolismo , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Estructura Terciaria de Proteína
12.
Cancer Lett ; 393: 16-21, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28188816

RESUMEN

Drug reprofiling is emerging as an effective paradigm for discovery of cancer treatments. Herein, an antipsychotic drug is immobilised using the Magic Tag® chemical genomics tool and screened against a T7 bacteriophage displayed library of polypeptides from Drosophila melanogaster, as a whole genome model, to uncover an interaction with a section of 17-ß-HSD10, a proposed prostate cancer target. A computational study and enzyme inhibition assay with full length human 17-ß-HSD10 identifies risperidone as a drug reprofiling candidate. When formulated with rumenic acid, risperidone slows proliferation of PC3 prostate cancer cells in vitro and retards PC3 prostate cancer tumour growth in vivo in xenografts in mice, presenting an opportunity to reprofile risperidone as a cancer treatment.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , 3-Hidroxiacil-CoA Deshidrogenasas/antagonistas & inhibidores , Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Antipsicóticos/farmacología , Drosophila melanogaster/efectos de los fármacos , Reposicionamiento de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Genómica/métodos , Neoplasias de la Próstata/tratamiento farmacológico , Risperidona/farmacología , 17-Hidroxiesteroide Deshidrogenasas/química , 17-Hidroxiesteroide Deshidrogenasas/genética , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/genética , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Animales , Antineoplásicos/química , Antipsicóticos/química , Bacteriófago T7/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Composición de Medicamentos , Inhibidores Enzimáticos/química , Biblioteca de Genes , Humanos , Ácidos Linoleicos Conjugados/química , Masculino , Ratones Desnudos , Simulación del Acoplamiento Molecular , Terapia Molecular Dirigida , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Conformación Proteica , Risperidona/química , Relación Estructura-Actividad , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cell Chem Biol ; 23(6): 727-37, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27291402

RESUMEN

S-Nitrosoglutathione (GSNO) is an endogenous transnitrosation donor involved in S-nitrosation of a variety of cellular proteins, thereby regulating diverse protein functions. Quantitative proteomic methods are necessary to establish which cysteine residues are most sensitive to GSNO-mediated transnitrosation. Here, a competitive cysteine-reactivity profiling strategy was implemented to quantitatively measure the sensitivity of >600 cysteine residues to transnitrosation by GSNO. This platform identified a subset of cysteine residues with a high propensity for GSNO-mediated transnitrosation. Functional characterization of previously unannotated S-nitrosation sites revealed that S-nitrosation of a cysteine residue distal to the 3-hydroxyacyl-CoA dehydrogenase type 2 (HADH2) active site impaired catalytic activity. Similarly, S-nitrosation of a non-catalytic cysteine residue in the lysosomal aspartyl protease cathepsin D (CTSD) inhibited proteolytic activation. Together, these studies revealed two previously uncharacterized cysteine residues that regulate protein function, and established a chemical-proteomic platform with capabilities to determine substrate specificity of other cellular transnitrosation agents.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Catepsina D/química , Catepsina D/metabolismo , Proteómica , 3-Hidroxiacil-CoA Deshidrogenasas/aislamiento & purificación , Humanos , Células MCF-7 , Nitrosación
14.
Nucleic Acids Res ; 43(10): 5112-9, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25925575

RESUMEN

SDR5C1 is an amino and fatty acid dehydrogenase/reductase, moonlighting as a component of human mitochondrial RNase P, which is the enzyme removing 5'-extensions of tRNAs, an early and crucial step in tRNA maturation. Moreover, a subcomplex of mitochondrial RNase P catalyzes the N(1)-methylation of purines at position 9, a modification found in most mitochondrial tRNAs and thought to stabilize their structure. Missense mutations in SDR5C1 cause a disease characterized by progressive neurodegeneration and cardiomyopathy, called HSD10 disease. We have investigated the effect of selected mutations on SDR5C1's functions. We show that pathogenic mutations impair SDR5C1-dependent dehydrogenation, tRNA processing and methylation. Some mutations disrupt the homotetramerization of SDR5C1 and/or impair its interaction with TRMT10C, the methyltransferase subunit of the mitochondrial RNase P complex. We propose that the structural and functional alterations of SDR5C1 impair mitochondrial RNA processing and modification, leading to the mitochondrial dysfunction observed in HSD10 patients.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/genética , Acetil-CoA C-Acetiltransferasa/deficiencia , Errores Innatos del Metabolismo Lipídico/genética , Mitocondrias/enzimología , Mutación , Ribonucleasa P/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Acetil-CoA C-Acetiltransferasa/genética , Discinesias , Discapacidad Intelectual Ligada al Cromosoma X , Metiltransferasas/metabolismo , Mitocondrias/genética , Procesamiento Postranscripcional del ARN , ARN de Transferencia/metabolismo
15.
J Hum Genet ; 59(11): 609-14, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25231369

RESUMEN

2-Methyl-3-hydroxybutyryl-CoA dehydrogenase (2M3HBD) deficiency (HSD10 disease) is a rare inborn error of metabolism, and <30 cases have been reported worldwide. This disorder is typically characterized by progressive neurodegenerative disease from 6 to 18 months of age. Here, we report the first patient with this disorder in Asia, with atypical clinical presentation. A 6-year-old boy, who had been well, presented with severe ketoacidosis following a 5-day history of gastroenteritis. Urinary organic acid analysis showed elevated excretion of 2-methyl-3-hydroxybutyrate and tiglylglycine. He was tentatively diagnosed with ß-ketothiolase (T2) deficiency. However, repeated enzyme assays using lymphocytes showed normal T2 activity and no T2 mutation was found. Instead, a hemizygous c.460G>A (p.A154T) mutation was identified in the HSD17B10 gene. This mutation was not found in 258 alleles from Japanese subjects (controls). A normal level of the HSD17B10 protein was found by immunoblot analysis but no 2M3HBD enzyme activity was detected in enzyme assays using the patient's fibroblasts. These data confirmed that this patient was affected with HSD10 disease. He has had no neurological regression until now. His fibroblasts showed punctate and fragmented mitochondrial organization by MitoTracker staining and had relatively low respiratory chain complex IV activity to those of other complexes.


Asunto(s)
Acetil-CoA C-Acetiltransferasa/deficiencia , Errores Innatos del Metabolismo Lipídico/genética , Mutación Puntual , 3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/genética , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Acetil-CoA C-Acetiltransferasa/genética , Secuencia de Bases , Carnitina/análogos & derivados , Carnitina/sangre , Niño , Análisis Mutacional de ADN , Diagnóstico Diferencial , Discinesias , Fibroblastos/metabolismo , Glicina/análogos & derivados , Glicina/orina , Humanos , Hidroxibutiratos/orina , Immunoblotting , Errores Innatos del Metabolismo Lipídico/diagnóstico , Masculino , Discapacidad Intelectual Ligada al Cromosoma X , Mitocondrias/metabolismo , Modelos Moleculares , Estructura Terciaria de Proteína
16.
J Microbiol Biotechnol ; 24(12): 1636-43, 2014 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-25112316

RESUMEN

3-Hydroxybutyryl-CoA dehydrogenase is an enzyme that catalyzes the second step in the biosynthesis of n-butanol from acetyl-CoA, in which acetoacetyl-CoA is reduced to 3-hydroxybutyryl-CoA. To understand the molecular mechanisms of n-butanol biosynthesis, we determined the crystal structure of 3-hydroxybutyryl-CoA dehydrogenase from Clostridium butyricum (CbHBD). The monomer structure of CbHBD exhibits a two-domain topology, with N- and C-terminal domains, and the dimerization of the enzyme was mostly constituted at the C-terminal domain. The mode of cofactor binding to CbHBD was elucidated by determining the crystal structure of the enzyme in complex with NAD(+). We also determined the enzyme's structure in complex with its acetoacetyl-CoA substrate, revealing that the adenosine diphosphate moiety was not highly stabilized compared with the remainder of the acetoacetyl-CoA molecule. Using this structural information, we performed a series of sitedirected mutagenesis experiments on the enzyme, such as changing residues located near the substrate-binding site, and finally developed a highly efficient CbHBD K50A/K54A/L232Y triple mutant enzyme that exhibited approximately 5-fold higher enzyme activity than did the wild type. The increased enzyme activity of the mutant was confirmed by enzyme kinetic measurements. The highly efficient mutant enzyme should be useful for increasing the production rate of n-butanol.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Clostridium butyricum/enzimología , Mutación Missense , 1-Butanol/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/genética , Acetilcoenzima A/metabolismo , Acilcoenzima A/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , NAD/metabolismo , Oxidación-Reducción , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína
17.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 7): 955-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25005097

RESUMEN

The (S)-3-hydroxybutyryl-CoA dehydrogenase PaaH1 from Ralstonia eutropha (RePaaH1) is an enzyme used in the biosynthesis of n-butanol from acetyl-CoA by the reduction of acetoacetyl-CoA to (S)-3-hydroxybutyryl-CoA. The RePaaH1 protein was crystallized using the hanging-drop vapour-diffusion method in the presence of 1.4 M ammonium sulfate, 0.1 M sodium cacodylate pH 6.0, 0.2 M sodium chloride at 295 K. X-ray diffraction data were collected to a maximum resolution of 2.6 Šon a synchrotron beamline. The crystal belonged to space group P3221, with unit-cell parameters a=b=135.4, c=97.2 Å. With three molecules per asymmetric unit, the crystal volume per unit protein weight (VM) is 2.68 Å3 Da(-1), which corresponds to a solvent content of approximately 54.1%. The structure was solved by the single-wavelength anomalous dispersion method and refinement of the structure is in progress.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , Proteínas Bacterianas/química , Cupriavidus necator/química , 1-Butanol/química , 3-Hidroxiacil-CoA Deshidrogenasas/genética , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Acetilcoenzima A/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Clonación Molecular , Cristalización , Cristalografía por Rayos X , Cupriavidus necator/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
Biochem Biophys Res Commun ; 448(2): 163-8, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24792376

RESUMEN

3-Hydroxybutyryl-CoA dehydrogenase is an enzyme involved in the synthesis of the biofuel n-butanol by converting acetoacetyl-CoA to 3-hydroxybutyryl-CoA. To investigate the molecular mechanism of n-butanol biosynthesis, we determined crystal structures of the Ralstonia eutropha-derived 3-hydroxybutyryl-CoA dehydrogenase (RePaaH1) in complex with either its cofactor NAD(+) or its substrate acetoacetyl-CoA. While the biologically active structure is dimeric, the monomer of RePaaH1 comprises two separated domains with an N-terminal Rossmann fold and a C-terminal helical bundle for dimerization. In this study, we show that the cofactor-binding site is located on the Rossmann fold and is surrounded by five loops and one helix. The binding mode of the acetoacetyl-CoA substrate was found to be that the adenosine diphosphate moiety is not highly stabilized compared with the remainder of the molecule. Residues involved in catalysis and substrate binding were further confirmed by site-directed mutagenesis experiments, and kinetic properties of RePaaH1were examined as well. Our findings contribute to the understanding of 3-hydroxybutyryl-CoA dehydrogenase catalysis, and will be useful in enhancing the efficiency of n-butanol biosynthesis by structure based protein engineering.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Cupriavidus necator/enzimología , 3-Hidroxiacil-CoA Deshidrogenasas/genética , Acilcoenzima A/química , Acilcoenzima A/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , NAD/química , NAD/metabolismo , Conformación Proteica
19.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 4): 485-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24699745

RESUMEN

(S)-3-Hydroxybutyryl-CoA dehydrogenase from Clostridium butyricum (CbHBD) is an enzyme that catalyzes the second step in the biosynthesis of n-butanol from acetyl-CoA by the reduction of acetoacetyl-CoA to 3-hydroxybutyryl-CoA. The CbHBD protein was crystallized using the hanging-drop vapour-diffusion method in the presence of 2 M ammonium sulfate, 0.1 M CAPS pH 10.5, 0.2 M lithium sulfate at 295 K. X-ray diffraction data were collected to a maximum resolution of 2.3 Šon a synchrotron beamline. The crystal belonged to space group R3, with unit-cell parameters a = b = 148.5, c = 201.6 Å. With four molecules per asymmetric unit, the crystal volume per unit protein weight (VM) is 3.52 Å(3) Da(-1), which corresponds to a solvent content of approximately 65.04%. The structure was solved by the molecular-replacement method and refinement of the structure is in progress.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , 3-Hidroxiacil-CoA Deshidrogenasas/aislamiento & purificación , Clonación Molecular , Clostridium butyricum/enzimología , Cristalización/métodos , Cristalografía por Rayos X/métodos , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Proteínas Recombinantes/metabolismo
20.
PLoS One ; 9(4): e95965, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24763278

RESUMEN

3-Hydroxyacyl-CoA dehydrogenase (HAD, EC 1.1.1.35) is a homodimeric enzyme localized in the mitochondrial matrix, which catalyzes the third step in fatty acid ß-oxidation. The crystal structures of human HAD and subsequent complexes with cofactor/substrate enabled better understanding of HAD catalytic mechanism. However, numerous human diseases were found related to mutations at HAD dimerization interface that is away from the catalytic pocket. The role of HAD dimerization in its catalytic activity needs to be elucidated. Here, we solved the crystal structure of Caenorhabditis elegans HAD (cHAD) that is highly conserved to human HAD. Even though the cHAD mutants (R204A, Y209A and R204A/Y209A) with attenuated interactions on the dimerization interface still maintain a dimerization form, their enzymatic activities significantly decrease compared to that of the wild type. Such reduced activities are in consistency with the reduced ratios of the catalytic intermediate formation. Further molecular dynamics simulations results reveal that the alteration of the dimerization interface will increase the fluctuation of a distal region (a.a. 60-80) that plays an important role in the substrate binding. The increased fluctuation decreases the stability of the catalytic intermediate formation, and therefore the enzymatic activity is attenuated. Our study reveals the molecular mechanism about the essential role of the HAD dimerization interface in its catalytic activity via allosteric effects.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/química , Proteínas de Caenorhabditis elegans/química , 3-Hidroxiacil-CoA Deshidrogenasas/genética , Animales , Biocatálisis , Proteínas de Caenorhabditis elegans/genética , Simulación de Dinámica Molecular , Mutación , Multimerización de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...