Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FEBS J ; 288(19): 5708-5722, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33792206

RESUMEN

Evolutionary robustness requires that the number of highly conserved amino acid residues in proteins is minimized. In enzymes, such conservation is observed for catalytic residues but also for some residues in the second shell or even further from the active site. ß-Lactamases evolve in response to changing antibiotic selection pressures and are thus expected to be evolutionarily robust, with a limited number of highly conserved amino acid residues. As part of the effort to understand the roles of conserved residues in class A ß-lactamases, we investigate the reasons leading to the conservation of two amino acid residues in the ß-lactamase BlaC, Glu37, and Trp229. Using site-directed mutagenesis, we have generated point mutations of these residues and observed a drastic decrease in the levels of soluble protein produced in Escherichia coli, thus abolishing completely the resistance of bacteria against ß-lactam antibiotics. However, the purified proteins are structurally and kinetically very similar to the wild-type enzyme, only differing by exhibiting a slightly lower melting temperature. We conclude that conservation of Glu37 and Trp229 is solely caused by an essential role in the folding process, and we propose that during folding Glu37 primes the formation of the central ß-sheet and Trp229 contributes to the hydrophobic collapse into a molten globule. ENZYME: EC 3.5.2.6. DATABASE: Structural data are available in PDB database under the accession number 7A5U.


Asunto(s)
Farmacorresistencia Bacteriana/genética , Conformación Proteica , Pliegue de Proteína/efectos de los fármacos , beta-Lactamasas/genética , Secuencia de Aminoácidos/genética , Sustitución de Aminoácidos/genética , Antibacterianos/efectos adversos , Antibacterianos/química , Antibacterianos/uso terapéutico , Dominio Catalítico/genética , Secuencia Conservada/genética , Escherichia coli/química , Escherichia coli/enzimología , Humanos , Cinética , Mutagénesis Sitio-Dirigida , beta-Lactamasas/ultraestructura
2.
Int J Mol Sci ; 22(6)2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33809335

RESUMEN

The relationship between protein motions (i.e., dynamics) and enzymatic function has begun to be explored in ß-lactamases as a way to advance our understanding of these proteins. In a recent study, we analyzed the dynamic profiles of TEM-1 (a ubiquitous class A ß-lactamase) and several ancestrally reconstructed homologues. A chief finding of this work was that rigid residues that were allosterically coupled to the active site appeared to have profound effects on enzyme function, even when separated from the active site by many angstroms. In the present work, our aim was to further explore the implications of protein dynamics on ß-lactamase function by altering the dynamic profile of TEM-1 using computational protein design methods. The Rosetta software suite was used to mutate amino acids surrounding either rigid residues that are highly coupled to the active site or to flexible residues with no apparent communication with the active site. Experimental characterization of ten designed proteins indicated that alteration of residues surrounding rigid, highly coupled residues, substantially affected both enzymatic activity and stability; in contrast, native-like activities and stabilities were maintained when flexible, uncoupled residues, were targeted. Our results provide additional insight into the structure-function relationship present in the TEM family of ß-lactamases. Furthermore, the integration of computational protein design methods with analyses of protein dynamics represents a general approach that could be used to extend our understanding of the relationship between dynamics and function in other enzyme classes.


Asunto(s)
Proteínas Mutantes/genética , Conformación Proteica , Ingeniería de Proteínas , beta-Lactamasas/genética , Aminoácidos/genética , Bacterias/enzimología , Sitios de Unión/genética , Dominio Catalítico/genética , Biología Computacional , Estabilidad de Enzimas/genética , Escherichia coli/enzimología , Modelos Moleculares , Simulación de Dinámica Molecular , Proteínas Mutantes/ultraestructura , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , beta-Lactamasas/ultraestructura
3.
J Struct Biol ; 211(2): 107544, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32512156

RESUMEN

The expression of ß-lactamases is a major mechanism of bacterial resistance to the ß-lactam antibiotics. Four molecular classes of ß-lactamases have been described (A, B, C and D), however until recently the class D enzymes were thought to exist only in Gram-negative bacteria. In the last few years, class D enzymes have been discovered in several species of Gram-positive microorganisms, such as Bacillus and Clostridia, and an investigation of their kinetic and structural properties has begun in earnest. Interestingly, it was observed that some species of Bacillus produce two distinct class D ß-lactamases, one highly active and the other with only basal catalytic activity. Analysis of amino acid sequences of active (BPU-1 from Bacillus pumilus) and inactive (BSU-2 from Bacillus subtilis and BAT-2 from Bacillus atrophaeus) enzymes suggests that presence of three additional amino acid residues in one of the surface loops of inefficient ß-lactamases may be responsible for their severely diminished activity. Our structural and docking studies show that the elongated loop of these enzymes severely restricts binding of substrates. Deletion of the three residues from the loops of BSU-2 and BAT-2 ß-lactamases relieves the steric hindrance and results in a significant increase in the catalytic activity of the enzymes. These data show that this surface loop plays an important role in modulation of the catalytic activity of Bacillus class D ß-lactamases.


Asunto(s)
Antibacterianos/química , Farmacorresistencia Bacteriana/genética , Conformación Proteica , beta-Lactamasas/ultraestructura , Secuencia de Aminoácidos/genética , Bacillus pumilus/efectos de los fármacos , Bacillus pumilus/enzimología , Bacillus subtilis/enzimología , Dominio Catalítico/genética , Clostridiaceae/enzimología , Cristalografía por Rayos X , Bacterias Gramnegativas/enzimología , Bacterias Gramnegativas/ultraestructura , Humanos , Simulación del Acoplamiento Molecular , Propiedades de Superficie , beta-Lactamasas/química , beta-Lactamasas/genética
4.
Indian J Med Res ; 144(1): 21-31, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27834322

RESUMEN

Klebsiella pneumoniae carbapenemases (KPCs) are plasmid encoded carbapenem hydrolyzing enzymes which have the potential to spread widely through gene transfer. The instability of upstream region of blaKPC accelerates emergence of different isoforms. Routine antibiotic susceptibility testing failed to detect KPC producers and some commercial kits have been launched for early identification of KPC producers. Notable among the drugs under development against KPC are mostly derivatives of polymixin; ß-lactamase inhibitor NXL104 with combination of oxyimino cephalosporin as well as with ceftazidime; a novel tricyclic carbapenem, LK-157, potentially useful against class A and class C enzymes; BLI-489-a bicyclic penem derivative; PTK-0796, a tetracycline derivative and ACHN-490. Combination therapy might be preferable to control KPC infections in immediate future. Clinicians are likely to opt for unconventional combinations of antibiotics to treat KPC infections because of unavailability of alternative agents. The KPCs have become endemic in many countries but there is no optimal treatment recommendation available for bacteria expressing KPCs. Reports of outbreaks involving KPCs have focused mainly on laboratory identification, empirical treatment outcomes and molecular epidemiology. This review includes information on the emergence of KPC variants, limitations of phenotyping methods, available molecular methods for identification of the KPC variants and treatment options highlighting the drugs under development.


Asunto(s)
Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Infecciones por Klebsiella/epidemiología , Klebsiella pneumoniae/genética , beta-Lactamasas/genética , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/ultraestructura , Carbapenémicos/uso terapéutico , Humanos , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/patogenicidad , Lactamas/uso terapéutico , Pruebas de Sensibilidad Microbiana , Filogenia , beta-Lactamasas/clasificación , beta-Lactamasas/ultraestructura
5.
PLoS Comput Biol ; 12(6): e1004926, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27332861

RESUMEN

Beta-lactamases represent the main bacterial mechanism of resistance to beta-lactam antibiotics and are a significant challenge to modern medicine. We have developed an automated classification and analysis protocol that exploits structure- and sequence-based approaches and which allows us to propose a grouping of serine beta-lactamases that more consistently captures and rationalizes the existing three classification schemes: Classes, (A, C and D, which vary in their implementation of the mechanism of action); Types (that largely reflect evolutionary distance measured by sequence similarity); and Variant groups (which largely correspond with the Bush-Jacoby clinical groups). Our analysis platform exploits a suite of in-house and public tools to identify Functional Determinants (FDs), i.e. residue sites, responsible for conferring different phenotypes between different classes, different types and different variants. We focused on Class A beta-lactamases, the most highly populated and clinically relevant class, to identify FDs implicated in the distinct phenotypes associated with different Class A Types and Variants. We show that our FunFHMMer method can separate the known beta-lactamase classes and identify those positions likely to be responsible for the different implementations of the mechanism of action in these enzymes. Two novel algorithms, ASSP and SSPA, allow detection of FD sites likely to contribute to the broadening of the substrate profiles. Using our approaches, we recognise 151 Class A types in UniProt. Finally, we used our beta-lactamase FunFams and ASSP profiles to detect 4 novel Class A types in microbiome samples. Our platforms have been validated by literature studies, in silico analysis and some targeted experimental verification. Although developed for the serine beta-lactamases they could be used to classify and analyse any diverse protein superfamily where sub-families have diverged over both long and short evolutionary timescales.


Asunto(s)
Algoritmos , Simulación del Acoplamiento Molecular/métodos , Análisis de Secuencia de Proteína/métodos , Programas Informáticos , beta-Lactamasas/química , beta-Lactamasas/ultraestructura , Sitios de Unión , Simulación por Computador , Farmacorresistencia Bacteriana , Activación Enzimática , Unión Proteica , Serina , Relación Estructura-Actividad , Especificidad por Sustrato , Resistencia betalactámica , Inhibidores de beta-Lactamasas/química
6.
J Biotechnol ; 235: 11-23, 2016 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-27046068

RESUMEN

The hydrolysis of amide bonds is a ubiquitous process in nature and is catalyzed by various enzymes: Whereas N-unsubstituted amides are cleaved by amidases (EC 3.5.1.4), peptidases (EC 3.4.X.X) cleave peptide bonds in proteins and are involved in a number of vital physiological processes. Cyclic amides (lactams) are generally not hydrolyzed by proteases, but require specific lactamases. While the ß-lactamase family (EC 3.5.2.6), acting on highly strained ß-lactams, is constantly growing, lactamases able to hydrolyze γ- and δ-lactams are largely under-represented, owing to the lack of ring strain of 5- and 6-membered cyclic amides which accounts for their lower reactivity. To date, the only known substrate in which a 5- or 6-membered ring lactam is enzymatically cleaved is (±)-2-azabicyclo[2.2.1]hept-5-en-3-one (rac-Vince lactam), as well as four derivatives thereof. For these industrially relevant substrates, enantiocomplementary biocatalysts have been identified and their stereopreference was found to correlate with their amino acid sequence and protein structure: While (+)-lactamases belong to the amidase signature family, displaying the typical GGSS(S/G)GS motif in the center of the protein sequence and a conserved Ser-Ser-Lys catalytic triad, (-)-lactamase activity has been identified only among serine hydrolases, members of the α/ß-hydrolase fold family, possessing a typical Ser-His-Asp catalytic triad. For larger 8- to 13-membered ring lactams, few active proteins have been identified, all are members of the amidase signature family. An enhanced partial CN double bond character in the amide bond explains the lower reactivity of particularly chemically stable lactams.


Asunto(s)
beta-Lactamasas , Hidrólisis , Lactamas/química , Lactamas/metabolismo , Modelos Moleculares , Estereoisomerismo , beta-Lactamasas/química , beta-Lactamasas/clasificación , beta-Lactamasas/metabolismo , beta-Lactamasas/ultraestructura
7.
Antimicrob Agents Chemother ; 60(2): 717-28, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26525784

RESUMEN

Class C ß-lactamases poorly hydrolyze cephamycins (e.g., cefoxitin, cefotetan, and moxalactam). In the past 2 decades, a new family of plasmid-based AmpC ß-lactamases conferring resistance to cefoxitin, the FOX family, has grown to include nine unique members descended from the Aeromonas caviae chromosomal AmpC. To understand the basis for the unique cephamycinase activity in the FOX family, we determined the first X-ray crystal structures of FOX-4, apo enzyme and the acyl-enzyme with its namesake compound, cefoxitin, using the Y150F deacylation-deficient variant. Notably, recombinant expression of N-terminally tagged FOX-4 also yielded an inactive adenylylated enzyme form not previously observed in ß-lactamases. The posttranslational modification (PTM), which occurs on the active site Ser64, would not seem to provide a selective advantage, yet might present an opportunity for the design of novel antibacterial drugs. Substantial ligand-induced changes in the enzyme are seen in the acyl-enzyme complex, particularly the R2 loop and helix H10 (P289 to N297), with movement of F293 by 10.3 Å. Taken together, this study provides the first picture of this highly proficient class C cephamycinase, uncovers a novel PTM, and suggests a possible cephamycin resistance mechanism involving repositioning of the substrate due to the presence of S153P, N289P, and N346I substitutions in the ligand binding pocket.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/ultraestructura , Cefoxitina/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Proteínas de Escherichia coli/ultraestructura , beta-Lactamasas/ultraestructura , Aeromonas caviae/efectos de los fármacos , Secuencia de Aminoácidos , Antibacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cefoxitina/metabolismo , Cristalografía por Rayos X , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/ultraestructura , Procesamiento Proteico-Postraduccional , Alineación de Secuencia , Espectrometría de Masas en Tándem , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
8.
Antimicrob Agents Chemother ; 60(2): 777-88, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26596949

RESUMEN

Around the world, Burkholderia spp. are emerging as pathogens highly resistant to ß-lactam antibiotics, especially ceftazidime. Clinical variants of Burkholderia pseudomallei possessing the class A ß-lactamase PenI with substitutions at positions C69 and P167 are known to demonstrate ceftazidime resistance. However, the biochemical basis for ceftazidime resistance in class A ß-lactamases in B. pseudomallei is largely undefined. Here, we performed site saturation mutagenesis of the C69 position and investigated the kinetic properties of the C69F variant of PenI from B. pseudomallei that results in a high level of ceftazidime resistance (2 to 64 mg/liter) when expressed in Escherichia coli. Surprisingly, quantitative immunoblotting showed that the steady-state protein levels of the C69F variant ß-lactamase were ∼4-fold lower than those of wild-type PenI (0.76 fg of protein/cell versus 4.1 fg of protein/cell, respectively). However, growth in the presence of ceftazidime increases the relative amount of the C69F variant to greater than wild-type PenI levels. The C69F variant exhibits a branched kinetic mechanism for ceftazidime hydrolysis, suggesting there are two different conformations of the enzyme. When incubated with an anti-PenI antibody, one conformation of the C69F variant rapidly hydrolyzes ceftazidime and most likely contributes to the higher levels of ceftazidime resistance observed in cell-based assays. Molecular dynamics simulations suggest that the electrostatic characteristics of the oxyanion hole are altered in the C69F variant. When ceftazidime was positioned in the active site, the C69F variant is predicted to form a greater number of hydrogen-bonding interactions than PenI with ceftazidime. In conclusion, we propose "a new twist" for enhanced ceftazidime resistance mediated by the C69F variant of the PenI ß-lactamase based on conformational changes in the C69F variant. Our findings explain the biochemical basis of ceftazidime resistance in B. pseudomallei, a pathogen of considerable importance, and suggest that the full repertoire of conformational states of a ß-lactamase profoundly affects ß-lactam resistance.


Asunto(s)
Antibacterianos/farmacología , Burkholderia pseudomallei/genética , Ceftazidima/farmacología , Escherichia coli/genética , Resistencia betalactámica/genética , beta-Lactamasas/genética , Burkholderia pseudomallei/efectos de los fármacos , Dominio Catalítico/genética , Cristalografía por Rayos X , Escherichia coli/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , beta-Lactamasas/ultraestructura
9.
Antimicrob Agents Chemother ; 59(8): 5069-72, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26055361

RESUMEN

We solved the crystal structure of the class C ß-lactamase MOX-1 complexed with the inhibitor aztreonam at 1.9Å resolution. The main-chain oxygen of Ser315 interacts with the amide nitrogen of aztreonam. Surprisingly, compared to that in the structure of free MOX-1, this main-chain carboxyl changes its position significantly upon binding to aztreonam. This result indicates that the interaction between MOX-1 and ß-lactams can be accompanied by conformational changes in the B3 ß-strand main chain.


Asunto(s)
Aztreonam/química , Proteínas Bacterianas/ultraestructura , Dominio Catalítico , Moxalactam/antagonistas & inhibidores , beta-Lactamasas/ultraestructura , Secuencia de Aminoácidos , Antibacterianos/farmacología , Aztreonam/farmacología , Proteínas Bacterianas/genética , Sitios de Unión , Dióxido de Carbono/química , Cristalografía por Rayos X , Farmacorresistencia Bacteriana Múltiple/genética , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Moxalactam/química , Moxalactam/farmacología , Conformación Proteica , Especificidad por Sustrato , beta-Lactamasas/genética
10.
Antimicrob Agents Chemother ; 59(7): 3700-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25691639

RESUMEN

ß-Lactamase enzymes (EC 3.5.2.6) are a significant threat to the continued use of ß-lactam antibiotics to treat infections. A novel non-ß-lactam ß-lactamase inhibitor with activity against many class A and C and some class D ß-lactamase variants, avibactam, is now available in the clinic in partnership with ceftazidime. Here, we explored the activity of avibactam against a variety of characterized isogenic laboratory constructs of ß-lactamase inhibitor-resistant variants of the class A enzyme SHV (M69I/L/V, S130G, K234R, R244S, and N276D). We discovered that the S130G variant of SHV-1 shows the most significant resistance to inhibition by avibactam, based on both microbiological and biochemical characterizations. Using a constant concentration of 4 mg/liter of avibactam as a ß-lactamase inhibitor in combination with ampicillin, the MIC increased from 1 mg/liter for blaSHV-1 to 256 mg/liter for blaSHV S130G expressed in Escherichia coli DH10B. At steady state, the k2/K value of the S130G variant when inactivated by avibactam was 1.3 M(-1) s(-1), versus 60,300 M(-1) s(-1) for the SHV-1 ß-lactamase. Under timed inactivation conditions, we found that an approximately 1,700-fold-higher avibactam concentration was required to inhibit SHV S130G than the concentration that inhibited SHV-1. Molecular modeling suggested that the positioning of amino acids in the active site of SHV may result in an alternative pathway of inactivation when complexed with avibactam, compared to the structure of CTX-M-15-avibactam, and that S130 plays a role in the acylation of avibactam as a general acid/base. In addition, S130 may play a role in recyclization. As a result, we advance that the lack of a hydroxyl group at position 130 in the S130G variant of SHV-1 substantially slows carbamylation of the ß-lactamase by avibactam by (i) removing an important proton acceptor and donator in catalysis and (ii) decreasing the number of H bonds. In addition, recyclization is most likely also slow due to the lack of a general base to initiate the process. Considering other inhibitor-resistant mechanisms among class A ß-lactamases, S130 may be the most important amino acid for the inhibition of class A ß-lactamases, perhaps even for the novel diazabicyclooctane class of ß-lactamase inhibitors.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Farmacorresistencia Bacteriana/fisiología , Escherichia coli/efectos de los fármacos , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/química , Acilación , Ampicilina/farmacología , Antibacterianos/farmacología , Dominio Catalítico/efectos de los fármacos , Ceftazidima/farmacología , Combinación de Medicamentos , Escherichia coli/genética , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , beta-Lactamasas/efectos de los fármacos , beta-Lactamasas/ultraestructura
11.
Antimicrob Agents Chemother ; 59(7): 3710-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25666153

RESUMEN

KPC-2 is the most prevalent class A carbapenemase in the world. Previously, KPC-2 was shown to hydrolyze the ß-lactamase inhibitors clavulanic acid, sulbactam, and tazobactam. In addition, substitutions at amino acid position R220 in the KPC-2 ß-lactamase increased resistance to clavulanic acid. A novel bridged diazabicyclooctane (DBO) non-ß-lactam ß-lactamase inhibitor, avibactam, was shown to inactivate the KPC-2 ß-lactamase. To better understand the mechanistic basis for inhibition of KPC-2 by avibactam, we tested the potency of ampicillin-avibactam and ceftazidime-avibactam against engineered variants of the KPC-2 ß-lactamase that possessed single amino acid substitutions at important sites (i.e., Ambler positions 69, 130, 234, 220, and 276) that were previously shown to confer inhibitor resistance in TEM and SHV ß-lactamases. To this end, we performed susceptibility testing, biochemical assays, and molecular modeling. Escherichia coli DH10B carrying KPC-2 ß-lactamase variants with the substitutions S130G, K234R, and R220M demonstrated elevated MICs for only the ampicillin-avibactam combinations (e.g., 512, 64, and 32 mg/liter, respectively, versus the MICs for wild-type KPC-2 at 2 to 8 mg/liter). Steady-state kinetics revealed that the S130G variant of KPC-2 resisted inactivation by avibactam; the k2/K ratio was significantly lowered 4 logs for the S130G variant from the ratio for the wild-type enzyme (21,580 M(-1) s(-1) to 1.2 M(-1) s(-1)). Molecular modeling and molecular dynamics simulations suggested that the mobility of K73 and its ability to activate S70 (i.e., function as a general base) may be impaired in the S130G variant of KPC-2, thereby explaining the slowed acylation. Moreover, we also advance the idea that the protonation of the sulfate nitrogen of avibactam may be slowed in the S130G variant, as S130 is the likely proton donor and another residue, possibly K234, must compensate. Our findings show that residues S130 as well as K234 and R220 contribute significantly to the mechanism of avibactam inactivation of KPC-2. Fortunately, the emergence of S130G, K234R, and R220M variants of KPC in the clinic should not result in failure of ceftazidime-avibactam, as the ceftazidime partner is potent against E. coli DH10B strains possessing all of these variants.


Asunto(s)
Ampicilina/farmacología , Compuestos de Azabiciclo/farmacología , Ceftazidima/farmacología , Escherichia coli/efectos de los fármacos , beta-Lactamasas/genética , Acilación , Sustitución de Aminoácidos/genética , Antibacterianos/farmacología , Cristalografía por Rayos X , Combinación de Medicamentos , Escherichia coli/enzimología , Escherichia coli/genética , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Simulación de Dinámica Molecular , Resistencia betalactámica/efectos de los fármacos , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/efectos de los fármacos , beta-Lactamasas/ultraestructura
12.
Appl Biochem Biotechnol ; 174(5): 1784-94, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25149455

RESUMEN

Streptomyces sp. isolate ERI-26 was obtained from the Nilgiris forest soil of Western Ghats, Tamil Nadu, India. Novel anthraquinone compound was isolated from the active fraction 5; it was identified by spectroscopical data using UV, IR, NMR and MASS. The isolated compound 1,5,7-trihydroxy-3-hydroxy methyl anthraquinone was tested against bacteria and fungi at minimum inhibitory concentration level. The compound showed significant antimicrobial activity against bacteria, Staphylococcus aureus at 125 µg/ml, Staphylococcus epidermidis at 62.5 µg/m, Bacillus subtilis at 31.25 µg/ml, fungi; Epidermophyton floccosum at 62.5 µg/ml, Aspergillus niger at 31.25 µg/ml, Aspergiller flavus at 31.25 µg/ml, Trichophyton rubrum at 62.5 µg/ml and Botrytis cinerea at 62.5 µg/ml. The isolated compound was subjected to molecular docking studies for the inhibition of TtgR, topoisomerase IV and AmpC ß-lactamase enzymes which are targets for antimicrobials. Docking studies of the compound showed low docking energy indicating its usefulness as antimicrobial agent. 1,5,7-Trihydroxy-3-hydroxy methyl anthraquinone is new, and its antimicrobial and molecular docking properties are reported for the first time.


Asunto(s)
Antraquinonas/química , Antraquinonas/farmacología , Fenómenos Fisiológicos Bacterianos/efectos de los fármacos , Proteínas Bacterianas/antagonistas & inhibidores , Topoisomerasa de ADN IV/antagonistas & inhibidores , Hongos/fisiología , Streptomyces/metabolismo , Antraquinonas/aislamiento & purificación , Antiinfecciosos , Proteínas Bacterianas/ultraestructura , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Topoisomerasa de ADN IV/ultraestructura , Hongos/efectos de los fármacos , Simulación del Acoplamiento Molecular , Unión Proteica , Microbiología del Suelo , Especificidad de la Especie , Streptomyces/clasificación , beta-Lactamasas/ultraestructura
13.
Int J Bioinform Res Appl ; 10(3): 235-63, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24794069

RESUMEN

ß-lactam antibiotics are utilised to treat bacterial infection. ß-lactamase enzymes (EC 3.5.2.6) are produced by several bacteria and are responsible for their resistance to ß-lactam antibiotics like penicillin, cephamycins and carbapenems. New Delhi Metallo-ß-lactamase (NDM-1) is a gene that makes bacteria resistant to ß-lactam antibiotics. Preparing a compound against NDM-1 will require additional investment and development by drug manufacturers as the current antibiotics will not treat patients with NDM-1 resistance. NDM-1 of Kolkata showed convergent-type evolution with other NDM-1 producing strains. The modelled structure exhibited α-ß-α barrel-type domain along with Zn metallo-ß-lactamase N-terminal domain. Compounds belonging to cephalosporins (relatively resistant to ß-lactamase) and other antibiotics ceftaroline, ceftobiprole, piperacillin, penamecillin, azidocillin, cefonicid, tigecycline and colistin have exhibited better binding affinity with the modelled NDM-1.


Asunto(s)
Antibacterianos/síntesis química , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Evolución Molecular , Modelos Químicos , beta-Lactamasas/química , beta-Lactamasas/genética , Sitios de Unión , Simulación por Computador , Inhibidores Enzimáticos/síntesis química , beta-Lactamasas/ultraestructura
14.
PLoS One ; 7(3): e31253, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22438863

RESUMEN

Nine neurodegenerative disorders, called polyglutamine (polyQ) diseases, are characterized by the formation of intranuclear amyloid-like aggregates by nine proteins containing a polyQ tract above a threshold length. These insoluble aggregates and/or some of their soluble precursors are thought to play a role in the pathogenesis. The mechanism by which polyQ expansions trigger the aggregation of the relevant proteins remains, however, unclear. In this work, polyQ tracts of different lengths were inserted into a solvent-exposed loop of the ß-lactamase BlaP and the effects of these insertions on the properties of BlaP were investigated by a range of biophysical techniques. The insertion of up to 79 glutamines does not modify the structure of BlaP; it does, however, significantly destabilize the enzyme. The extent of destabilization is largely independent of the polyQ length, allowing us to study independently the effects intrinsic to the polyQ length and those related to the structural integrity of BlaP on the aggregating properties of the chimeras. Only chimeras with 55Q and 79Q readily form amyloid-like fibrils; therefore, similarly to the proteins associated with diseases, there is a threshold number of glutamines above which the chimeras aggregate into amyloid-like fibrils. Most importantly, the chimera containing 79Q forms amyloid-like fibrils at the same rate whether BlaP is folded or not, whereas the 55Q chimera aggregates into amyloid-like fibrils only if BlaP is unfolded. The threshold value for amyloid-like fibril formation depends, therefore, on the structural integrity of the ß-lactamase moiety and thus on the steric and/or conformational constraints applied to the polyQ tract. These constraints have, however, no significant effect on the propensity of the 79Q tract to trigger fibril formation. These results suggest that the influence of the protein context on the aggregating properties of polyQ disease-associated proteins could be negligible when the latter contain particularly long polyQ tracts.


Asunto(s)
Amiloide/genética , Amiloide/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Péptidos/genética , Péptidos/metabolismo , Secuencia de Aminoácidos , Amiloide/química , Amiloide/ultraestructura , Bacillus/enzimología , Bacillus/genética , Cristalografía por Rayos X , Estabilidad de Enzimas , Humanos , Técnicas In Vitro , Cinética , Microscopía Electrónica de Transmisión , Modelos Moleculares , Datos de Secuencia Molecular , Enfermedades Neurodegenerativas/etiología , Péptidos/química , Conformación Proteica , Desnaturalización Proteica , Multimerización de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/ultraestructura , Termodinámica , Expansión de Repetición de Trinucleótido , beta-Lactamasas/química , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , beta-Lactamasas/ultraestructura
15.
J Struct Biol ; 173(1): 191-5, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20851187

RESUMEN

Proteins of the metallo-ß-lactamase (MßL) fold form a large superfamily of metallo-hydrolase/oxidoreductases. Members of this family are found in all three domains of life and are involved in a variety of biological functions related to hydrolysis, redox processes, DNA repair and uptake, and RNA processing. We classified the archaeal homologs of this superfamily based on sequence similarity and characterized a subfamily of the Cleavage and Polyadenylation Specificity Factor (CPSF) with an uncommon domain composition: in addition to an extended MßL domain, which accommodates the active site for RNA cleavage, this group has two N-terminal KH domains. Here, we present the crystal structure of a member of this group from Methanosarcina mazei. It reveals a dimerization mode of the MßL domain that has not been observed before and suggests that RNA is bound across the dimer interface, recognized by the KH domains of one monomer, and cleaved at the active site of the other.


Asunto(s)
Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Factor de Especificidad de Desdoblamiento y Poliadenilación/ultraestructura , Methanosarcina/enzimología , Modelos Moleculares , ARN/metabolismo , beta-Lactamasas/aislamiento & purificación , beta-Lactamasas/ultraestructura , Biología Computacional , Cristalización , Cartilla de ADN/genética , Dimerización
16.
J Biochem ; 147(6): 905-15, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20305272

RESUMEN

The X-ray crystal structure of metallo-beta-lactamase from Chryseobacterium indologenes IND-7 was determined at a resolution of 1.65 A. The overall structure adopted a four-layered alphabeta/betaalpha sandwich structure with a dinuclear zinc(II) active site, in which the zinc(II) ions were denoted as Zn1 and Zn2. The overall structure of IND-7 is analogous to those of subclass B1 metallo-beta-lactamases, as determined by X-ray crystallography. A significant structural difference, however, was observed in the dinuclear zinc(II) active site: the coordination geometry around Zn1 changed from tetrahedral, found in other subclass B1 metallo-beta-lactamases, to distorted trigonal bipyramidal, whereas that of Zn2 changed from trigonal bipyramidal to tetrahedral. Arg121(101), which is located in the vicinity of the dinuclear zinc(II) active site, may affect the binding affinity of Zn2 due to an electronic repulsion between the zinc(II) ion(s) and a positively charged guanidyl group of Arg121(101). Moreover, the hydrogen-bonding interaction of Arg121 with Ser71(53), which is conserved in IND-1, IND-3 and IND-5-IND-7, appeared to have important consequences for the binding affinity of Zn2 in conjunction with the above electrostatic effect.


Asunto(s)
Chryseobacterium/enzimología , beta-Lactamasas/ultraestructura , Sitios de Unión , Cristalografía por Rayos X , Enlace de Hidrógeno , Modelos Moleculares , Conformación Proteica , Homología de Secuencia de Aminoácido , Zinc/química , Zinc/metabolismo , beta-Lactamasas/química , beta-Lactamasas/aislamiento & purificación
17.
J Am Chem Soc ; 124(19): 5333-40, 2002 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11996574

RESUMEN

Although TEM-1 beta-lactamase is among the best studied enzymes, its acylation mechanism remains controversial. To investigate this problem, the structure of TEM-1 in complex with an acylation transition-state analogue was determined at ultrahigh resolution (0.85 A) by X-ray crystallography. The quality of the data was such as to allow for refinement to an R-factor of 9.1% and an R(free) of 11.2%. In the resulting structure, the electron density features were clear enough to differentiate between single and double bonds in carboxylate groups, to identify multiple conformations that are occupied by residues and loops, and to assign 70% of the protons in the protein. Unexpectedly, even at pH 8.0 where the protein was crystallized, the active site residue Glu166 is clearly protonated. This supports the hypothesis that Glu166 is the general base in the acylation half of the reaction cycle. This structure suggests that Glu166 acts through the catalytic water to activate Ser70 for nucleophilic attack on the beta-lactam ring of the substrate. The hydrolytic mechanism of class A beta-lactamases, such as TEM-1, appears to be symmetrical, as are the serine proteases. Apart from its mechanistic implications, this atomic resolution structure affords an unusually detailed view of the structure, dynamics, and hydrogen-bonding networks of TEM-1, which may be useful for the design of inhibitors against this key antibiotic resistance target.


Asunto(s)
Ácido Glutámico/química , beta-Lactamasas/química , Sitios de Unión , Cristalografía por Rayos X , Electrones , Hidrógeno/química , Modelos Moleculares , Conformación Proteica , beta-Lactamasas/metabolismo , beta-Lactamasas/ultraestructura
18.
J Mol Biol ; 234(1): 165-78, 1993 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-8230196

RESUMEN

The crystal structure of beta-lactamase from Staphylococcus aureus inactivated by p-nitrophenyl[[N-(benzyloxycarbonyl)amino]methyl]phosphonate, a methylphosphonate monoester monoanion inhibitor, has been determined and refined at 2.3 A resolution. The structure reveals a tetrahedral phosphorus covalently bonded to the O gamma atom of the active site serine, Ser70. One of the oxygen atoms bonded to phosphorus is located in the oxyanion hole formed by the two main-chain nitrogen atoms of Ser70 and Gln237, and the second bonded oxygen is solvated. The (benzyloxycarbonyl)aminomethyl group is oriented towards the active site gully such that the peptide group forms compensating electrostatic interactions with polar groups on the enzyme. The benzyl group forms a hydrophobic interaction with Ile239 and an aromatic-aromatic edge-to-face interaction with Tyr105, which has undergone a conformational transition relative to the native structure. The mode of binding supports the proposal that on reaction with the enzyme, the phosphonate generates a structure analogous to the tetrahedral transition state/intermediate associated with the acylation step of a normal substrate. The disposition of the phosphonyl group in this complex is the same as that of the corresponding phosphoryl group in the complex resulting from the inhibition of trypsin by diisopropylphosphofluoridate. The structure is consistent with a mechanism of inactivation that follows an associative pathway, proceeding via a transition state/intermediate in which phosphorus is penta-co-ordinated, forming a trigonal bipyramidal geometry with the phosphonyl donor (p-nitrophenol) and acceptor (Ser70 O gamma atom) in apical positions. A model of this transition state can be accommodated in the active site of beta-lactamase without any steric hindrance. A model of the tetrahedral transition state associated with the acylation step by benzyl penicillin has been derived. Because of the conformational rigidity of the fused rings of penicillin molecules, the orientation of the substrate is fixed once the tetrahedral carbonyl carbon and its ligands are superimposed on the phosphonate group. The outcome is that the carboxylate substituent on the thiazolidine ring forms a salt bridge with Lys234, and the preferred puckering of the ring is that observed in the crystal structure of ampicillin, the so-called "open" conformer.


Asunto(s)
Inhibidores de beta-Lactamasas , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Compuestos Organofosforados/química , Compuestos Organofosforados/farmacología , Penicilina G/química , Estructura Terciaria de Proteína , Serpinas/química , Staphylococcus aureus/enzimología , beta-Lactamasas/metabolismo , beta-Lactamasas/ultraestructura
19.
Nature ; 359(6397): 674, 1992 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-1436027
20.
Nature ; 359(6397): 700-5, 1992 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-1436034

RESUMEN

The X-ray crystal structure of the molecular complex of penicillin G with a deacylation-defective mutant of the RTEM-1 beta-lactamase from Escherichia coli shows how these antibiotics are recognized and destroyed. Penicillin G is covalently bound to Ser 70 0 gamma as an acyl-enzyme intermediate. The deduced catalytic mechanism uses Ser 70 0 gamma as the attacking nucleophile during acylation. Lys 73 N zeta acts as a general base in abstracting a proton from Ser 70 and transferring it to the thiazolidine ring nitrogen atom via Ser 130 0 gamma. Deacylation is accomplished by nucleophilic attack on the penicilloyl carbonyl carbon by a water molecule assisted by the general base, Glu 166.


Asunto(s)
Penicilina G , beta-Lactamasas/ultraestructura , Acilación , Secuencia de Aminoácidos , Sitios de Unión , Catálisis , Cristalografía , Escherichia coli/enzimología , Enlace de Hidrógeno , Modelos Moleculares , Datos de Secuencia Molecular , Penicilina G/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Serina/química , Relación Estructura-Actividad , Difracción de Rayos X , beta-Lactamasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...