Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Mol Biosci ; 10: 1111598, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36762206

RESUMEN

Irreversible inhibition of the enzyme type I dehydroquinase (DHQ1), a promising target for anti-virulence drug development, has been explored by enhancing the electrophilicity of specific positions of the ligand towards covalent lysine modification. For ligand design, we made use of the advantages offered by the intrinsic acid-base properties of the amino substituents introduced in the quinate scaffold, namely compounds 6-7 (R configuration at C3), to generate a potential leaving group, as well as the recognition pattern of the enzyme. The reactivity of the C2-C3 bond (Re face) in the scaffold was also explored using compound 8. The results of the present study show that replacement of the C3 hydroxy group of (-)-quinic acid by a hydroxyamino substituent (compound 6) provides a time-dependent irreversible inhibitor, while compound 7, in which the latter functionality was substituted by an amino group, and the introduction of an oxirane ring at C2-C3 bond, compound 8, do not allow covalent modification of the enzyme. These outcomes were supported by resolution of the crystal structures of DHQ1 from Staphylococcus aureus (Sa-DHQ1) and Salmonella typhi (St-DHQ1) chemically modified by 6 at a resolution of 1.65 and 1.90 Å, respectively, and of St-DHQ1 in the complex with 8 (1.55 Å). The combination of these structural studies with extensive molecular dynamics simulation studies allowed us to understand the molecular basis of the type of inhibition observed. This study is a good example of the importance of achieving the correct geometry between the reactive center of the ligand (electrophile) and the enzyme nucleophile (lysine residue) to allow selective covalent modification. The outcomes obtained with the hydroxyamino derivative 6 also open up new possibilities in the design of irreversible inhibitors based on the use of amino substituents.

2.
Chemistry ; 26(36): 8035-8044, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32259333

RESUMEN

Disabling the bacterial capacity to cause infection is an innovative approach that has attracted significant attention to fight against superbugs. A relevant target for anti-virulence drug discovery is the type I dehydroquinase (DHQ1) enzyme. It was shown that the 2-hydroxyethylammonium derivative 3 has in vitro activity since it causes the covalent modification of the catalytic lysine residue of DHQ1. As this compound does not bear reactive electrophilic centers, how the chemical modification occurs is intriguing. We report here an integrated approach, which involves biochemical studies, X-ray crystallography and computational studies on the reaction path using combined quantum mechanics/molecular mechanics Umbrella Sampling Molecular Dynamics, that evidences that DHQ1 catalyzes its self-immolation by transforming the unreactive 2-hydroxyethylammonium group in 3 into an epoxide that triggers the lysine covalent modification. This finding might open opportunities for the design of lysine-targeted irreversible inhibitors bearing a 2-hydroxyethylammonium moiety as an epoxide proform, which to our knowledge has not been reported previously.


Asunto(s)
Bacterias/química , Inhibidores Enzimáticos/química , Compuestos Epoxi/química , Hidroliasas/química , Bacterias/metabolismo , Catálisis , Descubrimiento de Drogas , Hidroliasas/metabolismo , Lisina , Simulación de Dinámica Molecular
3.
Nucleic Acids Res ; 46(1): 473-484, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29165717

RESUMEN

Bacterial biofilms are a complex architecture of cells that grow on moist interfaces, and are held together by a molecular glue of extracellular proteins, sugars and nucleic acids. Biofilms are particularly problematic in human healthcare as they can coat medical implants and are thus a potential source of disease. The enzymatic dispersal of biofilms is increasingly being developed as a new strategy to treat this problem. Here, we have characterized NucB, a biofilm-dispersing nuclease from a marine strain of Bacillus licheniformis, and present its crystal structure together with the biochemistry and a mutational analysis required to confirm its active site. Taken together, these data support the categorization of NucB into a unique subfamily of the ßßα metal-dependent non-specific endonucleases. Understanding the structure and function of NucB will facilitate its future development into an anti-biofilm therapeutic agent.


Asunto(s)
Bacillus licheniformis/fisiología , Proteínas Bacterianas/química , Biopelículas/crecimiento & desarrollo , Desoxirribonucleasas/química , Bacillus licheniformis/genética , Bacillus licheniformis/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , ADN/genética , ADN/metabolismo , Desoxirribonucleasas/genética , Desoxirribonucleasas/metabolismo , Modelos Moleculares , Conformación Proteica
4.
ChemMedChem ; 12(18): 1512-1524, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28791799

RESUMEN

A multidisciplinary approach was used to identify and optimize a quinazolinedione-based ligand that would decrease the flexibility of the substrate-covering loop (catalytic loop) of the type II dehydroquinase from Helicobacter pylori. This enzyme, which is essential for the survival of this bacterium, is involved in the biosynthesis of aromatic amino acids. A computer-aided fragment-based protocol (ALTA) was first used to identify the aromatic fragments able to block the interface pocket that separates two neighboring enzyme subunits and is located at the active site entrance. Chemical modification of its non-aromatic moiety through an olefin cross-metathesis and Seebach's self-reproduction of chirality synthetic principle allowed the development of a quinazolinedione derivative that disables the catalytic loop plasticity, which is essential for the enzyme's catalytic cycle. Molecular dynamics simulations revealed that the ligand would force the catalytic loop into an inappropriate arrangement for catalysis by strong interactions with the catalytic tyrosine and by expelling the essential arginine out of the active site.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/metabolismo , Hidroliasas/metabolismo , Simulación de Dinámica Molecular , Antibacterianos/síntesis química , Antibacterianos/química , Antibacterianos/metabolismo , Sitios de Unión , Dominio Catalítico , Inhibidores Enzimáticos/química , Helicobacter pylori/enzimología , Hidroliasas/antagonistas & inhibidores , Ligandos , Quinazolinonas/química , Quinazolinonas/metabolismo
5.
Arthritis Rheumatol ; 69(8): 1601-1611, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28464560

RESUMEN

OBJECTIVE: To assess the ability of matriptase, a type II transmembrane serine proteinase, to promote aggrecan loss from the cartilage of patients with osteoarthritis (OA) and to determine whether its inhibition can prevent aggrecan loss and cartilage damage in experimental OA. METHODS: Aggrecan release from human OA cartilage explants and human stem cell-derived cartilage discs was evaluated, and cartilage-conditioned media were used for Western blotting. Gene expression was analyzed by real-time polymerase chain reaction. Murine OA was induced by surgical destabilization of the medial meniscus, and matriptase inhibitors were administered via osmotic minipump or intraarticular injection. Cartilage damage was scored histologically and aggrecan cleavage was visualized immunohistochemically using specific neoepitope antibodies. RESULTS: The addition of soluble recombinant matriptase promoted a time-dependent release of aggrecan (and collagen) from OA cartilage, which was sensitive to metalloproteinase inhibition and protease-activated receptor 2 antagonism. Although engineered human (normal) cartilage discs failed to release aggrecan following matriptase addition, both matrix metalloproteinase- and aggrecanase-mediated cleavages of aggrecan were detected in human OA cartilage. Additionally, while matriptase did not directly degrade aggrecan, it promoted the accumulation of low-density lipoprotein receptor-related protein 1 (LRP-1) in conditioned media of the OA cartilage explants. Matriptase inhibition via neutralizing antibody or small molecule inhibitor significantly reduced cartilage damage scores in murine OA, which was associated with reduced generation of metalloproteinase-mediated aggrecan cleavage. CONCLUSION: Matriptase potently induces the release of metalloproteinase-generated aggrecan fragments as well as soluble LRP-1 from OA cartilage. Therapeutic targeting of matriptase proteolytic activity reduces metalloproteinase activity, further suggesting that this serine proteinase may have potential as a disease-modifying therapy in OA.


Asunto(s)
Agrecanos/efectos de los fármacos , Cartílago Articular/efectos de los fármacos , Osteoartritis de la Rodilla/metabolismo , Serina Endopeptidasas/farmacología , Proteína ADAMTS4/efectos de los fármacos , Proteína ADAMTS4/metabolismo , Proteína ADAMTS5/efectos de los fármacos , Proteína ADAMTS5/metabolismo , Anciano , Anciano de 80 o más Años , Agrecanos/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Western Blotting , Cartílago Articular/metabolismo , Cartílago Articular/patología , Endopeptidasas/efectos de los fármacos , Endopeptidasas/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Técnicas In Vitro , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/efectos de los fármacos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Metaloproteinasas de la Matriz/efectos de los fármacos , Metaloproteinasas de la Matriz/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Meniscos Tibiales/cirugía , Ratones , Persona de Mediana Edad , Osteoartritis de la Rodilla/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/farmacología , Serina Endopeptidasas/metabolismo
6.
Chemistry ; 22(50): 17988-18000, 2016 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-27699905

RESUMEN

Shikimate kinase (SK), the fifth enzyme of the aromatic amino acid biosynthesis, is a recognized target for antibiotic drug discovery. The potential of the distinct dynamic apolar gap, which isolates the natural substrate from the solvent environment for catalysis, and the motion of Mycobacterium tuberculosis and Helicobacter pylori SK enzymes, which was observed by molecular dynamics simulations, was explored for inhibition selectivity. The results of the biochemical and computational studies reveal that the incorporation of bulky groups at position C5 of 5-aminoshikimic acid and the natural substrate enhances the selectivity for the H. pylori enzyme due to key motion differences in the shikimic acid binding domain (mainly helix α5). These studies show that the less-exploited motion-based design approach not only is an alternative strategy for the development of competitive inhibitors, but could also be a way to achieve selectivity against a particular enzyme among its homologues.


Asunto(s)
Antibacterianos/química , Inhibidores Enzimáticos/síntesis química , Helicobacter pylori/enzimología , Mycobacterium tuberculosis/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Ácido Shikímico/análogos & derivados , Antibacterianos/farmacología , Inhibidores Enzimáticos/química , Congelación , Helicobacter pylori/química , Mycobacterium tuberculosis/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Ácido Shikímico/química
7.
J Med Chem ; 59(11): 5471-87, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27191386

RESUMEN

The large conformational changes observed by Molecular Dynamics simulation studies on the product release in the LID and shikimic acid binding (SB) domains of the shikimate kinase (SK) enzyme have been exploited in the development of reversible competitive inhibitors against SK from Mycobacterium tuberculosis and Helicobacter pylori. This enzyme is a recognized target for antibiotic drug discovery. The reported C5-substituted shikimic acid analogues interact with the dynamic apolar pocket that surrounds the C4 and C5 hydroxyl groups of the natural substrate, cause the opening of the LID and SB domains, and capture the essential arginine far from the ATP binding site as required for catalysis. The 3-nitrobenzyl 3e and 5-benzothiophenyl derivatives 3i proved to be the most potent inhibitors. An ester prodrug of 3i was the most efficient derivative in achieving good in vitro activity against H. pylori, having a MIC value of 4 µg/mL.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Helicobacter pylori/enzimología , Modelos Moleculares , Estructura Molecular , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Relación Estructura-Actividad
9.
Chemistry ; 22(8): 2758-68, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26797764

RESUMEN

The phosphoryl-transfer mechanism of shikimate kinase from Mycobacterium tuberculosis and Helicobacter pylori, which is an attractive target for antibiotic drug discovery, has been studied by 1D (1)H and (31)P NMR spectroscopy. Metaphosphoric acid proved to be a good mimetic of the metaphosphate intermediate and facilitated the ready and rapid evaluation by NMR spectroscopic analysis of a dissociative mechanism. The required closed form of the active site for catalysis was achieved by the use of ADP (product) or two synthetic ADP analogues (AMPNP, AMPCP). Molecular dynamics simulation studies reported here also revealed that the essential arginine (Arg116/Arg117 in H. pylori and M. tuberculosis, respectively), which activates the γ-phosphate group of ATP for catalysis and triggers the release of the product for turnover, would also be involved in the stabilisation of the metaphosphate intermediate during catalysis. We believe that the studies reported here will be helpful for future structure-based design of inhibitors of this attractive target. The approach is also expected be useful for studies on the possible dissociative mechanism of other kinase enzymes.


Asunto(s)
Adenosina Difosfato/química , Adenosina Trifosfato/análogos & derivados , Helicobacter pylori/enzimología , Mycobacterium tuberculosis/enzimología , Ácidos Fosforosos/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Adenosina Trifosfato/química , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Helicobacter pylori/química , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Mycobacterium tuberculosis/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo
10.
J Am Chem Soc ; 137(29): 9333-43, 2015 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-26148116

RESUMEN

The first example of an ammonium derivative that causes a specific modification of the active site of type I dehydroquinase (DHQ1), a dehydratase enzyme that is a promising target for antivirulence drug discovery, is described. The resolution at 1.35 Å of the crystal structure of DHQ1 from Salmonella typhi chemically modified by this ammonium derivative revealed that the ligand is covalently attached to the essential Lys170 through the formation of an amine. The detection by mass spectroscopy of the reaction intermediates, in conjunction with the results of molecular dynamics simulations, allowed us to explain the inhibition mechanism and the experimentally observed differences between S. typhi and Staphylococcus aureus enzymes. The results presented here reveal that the replacement of Phe225 in St-DHQ1 by Tyr214 in Sa-DHQ1 and its hydrogen bonding interaction with the conserved water molecule observed in several crystal structures protects the amino adduct against further dehydration/aromatization reactions. In contrast, for the St-DHQ1 enzyme, the carboxylate group of Asp114, with the assistance of this water molecule, would trigger the formation of a Schiff base that can undergo further dehydration reactions until full aromatization of the cyclohexane ring is achieved. Moreover, in vitro antivirulence studies showed that the reported compound is able to reduce the ability of Salmonella Enteritidis to kill A459 respiratory cells. These studies have identified a good scaffold for the design of irreversible inhibitors that can be used as drugs and has opened up new opportunities for the development of novel antivirulence agents by targeting the DHQ1 enzyme.


Asunto(s)
Compuestos de Amonio/química , Compuestos de Amonio/farmacología , Dominio Catalítico/efectos de los fármacos , Hidroliasas/antagonistas & inhibidores , Hidroliasas/química , Salmonella typhi/enzimología , Staphylococcus aureus/enzimología , Compuestos de Amonio/metabolismo , Línea Celular , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Hidroliasas/metabolismo , Simulación de Dinámica Molecular , Salmonella typhi/patogenicidad , Virulencia
11.
Nat Commun ; 6: 7481, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-26112186

RESUMEN

The structure of the human gut microbiota is controlled primarily through the degradation of complex dietary carbohydrates, but the extent to which carbohydrate breakdown products are shared between members of the microbiota is unclear. We show here, using xylan as a model, that sharing the breakdown products of complex carbohydrates by key members of the microbiota, such as Bacteroides ovatus, is dependent on the complexity of the target glycan. Characterization of the extensive xylan degrading apparatus expressed by B. ovatus reveals that the breakdown of the polysaccharide by the human gut microbiota is significantly more complex than previous models suggested, which were based on the deconstruction of xylans containing limited monosaccharide side chains. Our report presents a highly complex and dynamic xylan degrading apparatus that is fine-tuned to recognize the different forms of the polysaccharide presented to the human gut microbiota.


Asunto(s)
Bacteroides/metabolismo , Xilanos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bifidobacterium/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Genómica , Humanos , Transporte de Proteínas , Zea mays
12.
Org Biomol Chem ; 13(3): 706-16, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25370445

RESUMEN

The irreversible inhibition of type I dehydroquinase (DHQ1), the third enzyme of the shikimic acid pathway, is investigated by structural, biochemical and computational studies. Two epoxides, which are mimetics of the natural substrate, were designed as irreversible inhibitors of the DHQ1 enzyme and to study the binding requirements of the linkage to the enzyme. The epoxide with the S configuration caused the covalent modification of the protein whereas no reaction was obtained with its epimer. The first crystal structure of DHQ1 from Salmonella typhi covalently modified by the S epoxide, which is reported at 1.4 Å, revealed that the modified ligand is surprisingly covalently attached to the essential Lys170 by the formation of a stable Schiff base. The experimental and molecular dynamics simulation studies reported here highlight the huge importance of the conformation of the C3 carbon of the ligand for covalent linkage to this type of aldolase I enzyme, revealed the key role played by the essential His143 as a Lewis acid in this process and show the need for a neatly closed active site for catalysis.


Asunto(s)
Proteínas Bacterianas/química , Inhibidores Enzimáticos/química , Compuestos Epoxi/química , Hidroliasas/química , Bases de Schiff/química , Proteínas Bacterianas/antagonistas & inhibidores , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Compuestos Epoxi/síntesis química , Histidina/química , Hidroliasas/antagonistas & inhibidores , Enlace de Hidrógeno , Cinética , Ligandos , Lisina/química , Simulación de Dinámica Molecular , Unión Proteica , Salmonella typhi/química , Salmonella typhi/enzimología , Electricidad Estática
13.
Biochem J ; 462(3): 415-24, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24957267

RESUMEN

Structural, biochemical and computational studies to study substrate binding and the role of the conserved residues of the DHQ1 (type I dehydroquinase) enzyme active site are reported in the present paper. The crystal structure of DHQ1 from Salmonella typhi in complex with (2R)-2-methyl-3-dehydroquinic acid, a substrate analogue, was solved at 1.5 Å. The present study reveals a previously unknown key role for conserved Glu46, Phe145 and Met205 and Gln236, Pro234 and Ala233 residues, with the latter three being located in the flexible substrate-covering loop. Gln236 was shown to be responsible for the folding of this loop and for the dramatic reduction of its flexibility, which triggers active site closure. Glu46 was found to be key in bringing the substrate close to the lysine/histidine catalytic pocket to initiate catalysis. The present study could be useful in the rational design of inhibitors of this challenging and recognized target for the development of novel herbicides and antimicrobial agents.


Asunto(s)
Hidroliasas/metabolismo , Secuencia de Aminoácidos , Catálisis , Dominio Catalítico , Cristalización , Cristalografía por Rayos X , Cinética , Simulación de Dinámica Molecular , Salmonella typhi/enzimología , Relación Estructura-Actividad
14.
J Med Chem ; 57(8): 3494-510, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24689821

RESUMEN

Structural and computational studies to explore the WAT1 binding pocket in the structure-based design of inhibitors against the type II dehydroquinase (DHQ2) enzyme are reported. The crystal structures of DHQ2 from M. tuberculosis in complex with four of the reported compounds are described. The electrostatic interaction observed between the guanidinium group of the essential arginine and the carboxylate group of one of the inhibitors in the reported crystal structures supports the recently suggested role of this arginine as the residue that triggers the release of the product from the active site. The results of the structural and molecular dynamics simulation studies revealed that the inhibitory potency is favored by promoting interactions with WAT1 and the residues located within this pocket and, more importantly, by avoiding situations where the ligands occupy the WAT1 binding pocket. The new insights can be used to advantage in the structure-based design of inhibitors.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Hidroliasas/antagonistas & inhibidores , Agua/química , Cristalización , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Hidroliasas/química , Simulación de Dinámica Molecular , Relación Estructura-Actividad
15.
Biochem J ; 458(3): 547-57, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24392963

RESUMEN

DHQ2 (type II dehydroquinase), which is an essential enzyme in Helicobacter pylori and Mycobacterium tuberculosis and does not have any counterpart in humans, is recognized to be an attractive target for the development of new antibacterial agents. Computational and biochemical studies that help understand in atomic detail the catalytic mechanism of these bacterial enzymes are reported in the present paper. A previously unknown key role of certain conserved residues of these enzymes, as well as the structural changes responsible for triggering the release of the product from the active site, were identified. Asp89*/Asp88* from a neighbouring enzyme subunit proved to be the residue responsible for the deprotonation of the essential tyrosine to afford the catalytic tyrosinate, which triggers the enzymatic process. The essentiality of this residue is supported by results from site-directed mutagenesis. For H. pylori DHQ2, this reaction takes place through the assistance of a water molecule, whereas for M. tuberculosis DHQ2, the tyrosine is directly deprotonated by the aspartate residue. The participation of a water molecule in this deprotonation reaction is supported by solvent isotope effects and proton inventory studies. MD simulation studies provide details of the required motions for the catalytic turnover, which provides a complete overview of the catalytic cycle. The product is expelled from the active site by the essential arginine residue and after a large conformational change of a loop containing two conserved arginine residues (Arg109/Arg108 and Arg113/Arg112), which reveals a previously unknown key role for these residues. The present study highlights the key role of the aspartate residue whose blockage could be useful in the rational design of inhibitors and the mechanistic differences between both enzymes.


Asunto(s)
Proteínas Bacterianas/química , Helicobacter pylori/enzimología , Hidroliasas/química , Mycobacterium tuberculosis/enzimología , Arginina/química , Ácido Aspártico/química , Proteínas Bacterianas/genética , Catálisis , Hidroliasas/genética , Cinética , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Teoría Cuántica , Solventes
16.
J Am Chem Soc ; 135(33): 12366-76, 2013 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-23889343

RESUMEN

Shikimate kinase (SK) is an essential enzyme in several pathogenic bacteria and does not have any counterpart in human cells, thus making it an attractive target for the development of new antibiotics. The key interactions of the substrate and product binding and the enzyme movements that are essential for catalytic turnover of the Mycobacterium tuberculosis shikimate kinase enzyme (Mt-SK) have been investigated by structural and computational studies. Based on these studies several substrate analogs were designed and assayed. The crystal structure of Mt-SK in complex with ADP and one of the most potent inhibitors has been solved at 2.15 Å. These studies reveal that the fixation of the diaxial conformation of the C4 and C5 hydroxyl groups recognized by the enzyme or the replacement of the C3 hydroxyl group in the natural substrate by an amino group is a promising strategy for inhibition because it causes a dramatic reduction of the flexibility of the LID and shikimic acid binding domains. Molecular dynamics simulation studies showed that the product is expelled from the active site by three arginines (Arg117, Arg136, and Arg58). This finding represents a previously unknown key role of these conserved residues. These studies highlight the key role of the shikimic acid binding domain in the catalysis and provide guidance for future inhibitor designs.


Asunto(s)
Biocatálisis , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Mycobacterium tuberculosis/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Adenosina Difosfato/metabolismo , Dominio Catalítico , Inhibidores Enzimáticos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Ácido Shikímico/química , Ácido Shikímico/metabolismo
17.
ACS Chem Biol ; 8(3): 568-77, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23198883

RESUMEN

The structural changes caused by the substitution of the aromatic moiety in (2S)-2-benzyl-3-dehydroquinic acids and its epimers in C2 by electron-withdrawing or electron-donating groups in type II dehydroquinase enzyme from M. tuberculosis and H. pylori has been investigated by structural and computational studies. Both compounds are reversible competitive inhibitors of this enzyme, which is essential in these pathogenic bacteria. The crystal structures of M. tuberculosis and H. pylori in complex with (2S)-2-(4-methoxy)benzyl- and (2S)-2-perfluorobenzyl-3-dehydroquinic acids have been solved at 2.0, 2.3, 2.0, and 1.9 Å, respectively. The crystal structure of M. tuberculosis in complex with (2R)-2-(benzothiophen-5-yl)methyl-3-dehydroquinic acid is also reported at 1.55 Å. These crystal structures reveal key differences in the conformation of the flexible loop of the two enzymes, a difference that depends on the presence of electron-withdrawing or electron-donating groups in the aromatic moiety of the inhibitors. This loop closes over the active site after substrate binding, and its flexibility is essential for the function of the enzyme. These differences have also been investigated by molecular dynamics simulations in an effort to understand the significant inhibition potency differences observed between some of these compounds and also to obtain more information about the possible movements of the loop. These computational studies have also allowed us to identify key structural factors of the H. pylori loop that could explain its reduced flexibility in comparison to the M. tuberculosis loop, specifically by the formation of a key salt bridge between the side chains of residues Asp18 and Arg20.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hidroliasas/antagonistas & inhibidores , Ácido Quínico/análogos & derivados , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Helicobacter pylori/enzimología , Hidroliasas/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Estructura Molecular , Mycobacterium tuberculosis/enzimología , Ácido Quínico/síntesis química , Ácido Quínico/química , Ácido Quínico/farmacología
18.
FEBS Lett ; 586(6): 675-9, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22449962

RESUMEN

MbeA and MbeC are two key proteins in plasmid ColE1 conjugal mobilization. Isothermal titration calorimetry was used to detect and quantify an interaction between MbeA and MbeC. As a result of this interaction, the affinity of MbeA for single stranded DNA increased. The interaction was confirmed in vivo using a bacterial two-hybrid system, which revealed that MbeA-MbeC complexes are formed through the amino-terminal region of MbeA and the carboxy-terminal region of MbeC. To the best of our knowledge, this is the first report of direct interactions between conjugative proteins encoded by a mobilizable plasmid.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endodesoxirribonucleasas/metabolismo , Plásmidos/metabolismo , Proteínas Bacterianas/genética , Secuencia de Bases , Conjugación Genética , ADN de Cadena Simple/metabolismo , Endodesoxirribonucleasas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Datos de Secuencia Molecular , Plásmidos/genética , Técnicas del Sistema de Dos Híbridos
19.
Org Biomol Chem ; 10(18): 3662-76, 2012 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-22447158

RESUMEN

Several 3-alkylaryl mimics of the enol intermediate in the reaction catalyzed by type II dehydroquinase were synthesized to investigate the effect on the inhibition potency of replacing the oxygen atom in the side chain by a carbon atom. The length and the rigidity of the spacer was also studied. The inhibitory properties of the reported compounds against type II dehydroquinase from Mycobacterium tuberculosis and Helicobacter pylori are also reported. The binding modes of these analogs in the active site of both enzymes were studied by molecular docking using GOLD 5.0 and dynamic simulations studies.


Asunto(s)
Enoil-CoA Hidratasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Cetonas/farmacología , Imitación Molecular , Enoil-CoA Hidratasa/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Helicobacter pylori/enzimología , Cetonas/síntesis química , Cetonas/química , Modelos Moleculares , Simulación de Dinámica Molecular , Estructura Molecular , Mycobacterium tuberculosis/enzimología , Estereoisomerismo , Relación Estructura-Actividad
20.
J Med Chem ; 54(17): 6063-84, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21780742

RESUMEN

The synthesis of high-affinity reversible competitive inhibitors of Mycobacterium tuberculosis type II dehydroquinase, an essential enzyme in Mycobacterium tuberculosis bacteria, is reported. The inhibitors reported here are mimics of the enol intermediate and the effect of substitution on C2 was studied. The crystal structures of Mycobacterium tuberculosis type II dehydroquinase in complex with three of the reported inhibitors are also described. The results show that an aromatic substituent on C2 prevents the closure of the active site by impeding the hydrogen-bonding interaction of Arg108 with the essential Tyr24 of the flexible loop, the residue that initiates catalysis. Chemical modifications of the reported acids were also carried out to improve internalization into Mycobacterium tuberculosis through an ester prodrug approach. Propyl esters proved to be the most efficient in achieving optimal in vitro activities.


Asunto(s)
Antituberculosos/farmacología , Inhibidores Enzimáticos/farmacología , Hidroliasas/antagonistas & inhibidores , Mycobacterium tuberculosis/efectos de los fármacos , Profármacos/farmacología , Antituberculosos/síntesis química , Antituberculosos/química , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Mycobacterium tuberculosis/crecimiento & desarrollo , Oxazinas , Profármacos/síntesis química , Profármacos/química , Relación Estructura-Actividad , Xantenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...