Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Protein Sci ; 33(4): e4964, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38501584

RESUMEN

Worldwide, tuberculosis is the second leading infectious killer and multidrug resistance severely hampers disease control. Mycolic acids are a unique category of lipids that are essential for viability, virulence, and persistence of the causative agent, Mycobacterium tuberculosis (Mtb). Therefore, enzymes involved in mycolic acid biosynthesis represent an important class of drug targets. We previously showed that the (3R)-hydroxyacyl-ACP dehydratase (HAD) protein HadD is dedicated mainly to the production of ketomycolic acids and plays a determinant role in Mtb biofilm formation and virulence. Here, we discovered that HAD activity requires the formation of a tight heterotetramer between HadD and HadB, a HAD unit encoded by a distinct chromosomal region. Using biochemical, structural, and cell-based analyses, we showed that HadB is the catalytic subunit, whereas HadD is involved in substrate binding. Based on HadBDMtb crystal structure and substrate-bound models, we identified determinants of the ultra-long-chain lipid substrate specificity and revealed details of structure-function relationship. HadBDMtb unique function is partly due to a wider opening and a higher flexibility of the substrate-binding crevice in HadD, as well as the drastically truncated central α-helix of HadD hotdog fold, a feature described for the first time in a HAD enzyme. Taken together, our study shows that HadBDMtb , and not HadD alone, is the biologically relevant functional unit. These results have important implications for designing innovative antivirulence molecules to fight tuberculosis, as they suggest that the target to consider is not an isolated subunit, but the whole HadBD complex.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Acido Graso Sintasa Tipo II/química , Ácidos Micólicos/metabolismo , Hidroliasas/química
2.
FEBS J ; 289(16): 4963-4980, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35175661

RESUMEN

Comprehending the molecular strategies employed by Mycobacterium tuberculosis (Mtb) in FAS-II regulation is of paramount significance for curbing tuberculosis progression. Mtb employs two sets of dehydratases, namely HadAB and HadBC (ß-hydroxyacyl acyl carrier protein dehydratase), for the regulation of the fatty acid synthase (FAS-II) pathway. We utilized a sequence similarity network to discern the basis for the presence of two copies of the dehydratase gene in Mtb. This analysis groups HadC and HadA in different clusters, which could be attributed to the variability in their physiological role with respect to the acyl chain uptake. Our study reveals structural details pertaining to the crystal structure of the last remaining enzyme of the FAS-II pathway. It also provides insights into the highly flexible hot-dog helix and substrate regulatory loop. Additionally, mutational studies assisted in establishing the role of the C-terminal end in HadC of HadBC in the regulation of acyl carrier protein from Mtb-mediated interactions. Complemented with surface plasmon resonance and molecular dynamics simulation studies, the present study provides the first evidence of the molecular mechanisms involved in the differential binding affinity of the acyl carrier protein from Mtb towards both mtbHadAB and mtbHadBC.


Asunto(s)
Mycobacterium tuberculosis , Ácidos Micólicos , Proteína Transportadora de Acilo/genética , Proteína Transportadora de Acilo/metabolismo , Proteínas Bacterianas/metabolismo , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/genética , Acido Graso Sintasa Tipo II/metabolismo , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Hidroliasas/metabolismo , Mycobacterium tuberculosis/metabolismo , Ácidos Micólicos/metabolismo
3.
Med Chem ; 17(5): 474-484, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-31763975

RESUMEN

BACKGROUND: Antimicrobial resistance is a persistent problem regarding infection treatment and calls for developing new antimicrobial agents. Inhibition of bacterial ß-ketoacyl acyl carrier protein synthase III (FabH), which catalyzes the condensation reaction between a CoAattached acetyl group and an ACP-attached malonyl group in bacteria is an interesting strategy to find new antibacterial agents. OBJECTIVE: The aim of this work was to design and synthesize arylsulfonylhydrazones potentially FabH inhibitors and evaluate their antimicrobial activity. METHODS: MIC50 values of sulfonylhydrazones against E. coli and S. aureus were determined. Antioxidant activity was evaluated by DPPH (1-1'-diphenyl-2-picrylhydrazyl) assay and cytotoxicity against LL24 lung fibroblast cells was verified by MTT method. Principal component analysis (PCA) was performed in order to suggest a structure-activity relationship. Molecular docking allowed to propose sulfonylhydrazones interactions with FabH. RESULTS: The most active compound showed activity against S. aureus and E. coli, with MIC50 = 0.21 and 0.44 µM, respectively. PCA studies correlated better activity to lipophilicity and molecular docking indicated that sulfonylhydrazone moiety is important to hydrogen-bond with FabH while methylcatechol ring performs π-π stacking interaction. The DPPH assay revealed that some sulfonylhydrazones derived from the methylcatechol series had antioxidant activity. None of the evaluated compounds was cytotoxic to human lung fibroblast cells, suggesting that the compounds might be considered safe at the tested concentration. CONCLUSION: Arylsufonylhydrazones is a promising scaffold to be explored for the design of new antimicrobial agents.


Asunto(s)
3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/antagonistas & inhibidores , Antibacterianos/farmacología , Inhibidores Enzimáticos/farmacología , Hidrazonas/farmacología , Sulfonamidas/farmacología , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/química , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/metabolismo , Acetiltransferasas/antagonistas & inhibidores , Acetiltransferasas/química , Acetiltransferasas/metabolismo , Antibacterianos/síntesis química , Antibacterianos/metabolismo , Dominio Catalítico , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Escherichia coli/efectos de los fármacos , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Acido Graso Sintasa Tipo II/antagonistas & inhibidores , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/metabolismo , Hidrazonas/síntesis química , Hidrazonas/metabolismo , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Estructura Molecular , Análisis de Componente Principal , Unión Proteica , Staphylococcus aureus/efectos de los fármacos , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/metabolismo
4.
Biochemistry ; 59(50): 4735-4743, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33283513

RESUMEN

Long-chain polyunsaturated fatty acids (LC-PUFAs) are essential ingredients of the human diet. They are synthesized by LC-PUFA synthases (PFASs) expressed in marine bacteria and other organisms. PFASs are large enzyme complexes that are homologous to mammalian fatty acid synthases and microbial polyketide synthases. One subunit of each PFAS harbors consecutive ketosynthase (KSc) and chain length factor (CLF) domains that collectively catalyze the elongation of a nascent fatty acyl chain via iterative carbon-carbon bond formation. We report the X-ray crystal structure of the KS-CLF didomain from a well-studied PFAS in Moritella marina. Our structure, in combination with biochemical analysis, provides a foundation for understanding the mechanism of substrate recognition and chain length control by the KS-CLF didomain as well as its interaction with a cognate acyl carrier protein partner.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/metabolismo , Ácidos Grasos Insaturados/biosíntesis , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Vías Biosintéticas , Dominio Catalítico/genética , Cristalografía por Rayos X , Acido Graso Sintasa Tipo II/genética , Ácidos Grasos Insaturados/química , Humanos , Espectrometría de Masas , Modelos Moleculares , Moritella/enzimología , Moritella/genética , Mutagénesis Sitio-Dirigida , Dominios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Electricidad Estática , Especificidad por Sustrato
5.
Eur J Med Chem ; 208: 112757, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-32883635

RESUMEN

Development of new anti-bacterial agents acting upon underexploited targets and thus evading known mechanisms of resistance is the need of the hour. The highly conserved and distinct bacterial fatty acid biosynthesis pathway (FAS-II), presents a validated and yet relatively underexploited target for drug discovery. FabI and its isoforms (FabL, FabK, FabV and InhA) are essential enoyl-ACP reductases present in several microorganisms. In addition, the components of the FAS-II pathway are distinct from the multi-enzyme FAS-I complex found in mammals. Thus, inhibition of FabI and its isoforms is anticipated to result in broad-spectrum antibacterial activity. Several research groups from industry and academic laboratories have devoted significant efforts to develop effective FabI-targeting antibiotics, which are currently in various stages of clinical development for the treatment of multi-drug resistant bacterial infections. This review summarizes all the natural as well as synthetic inhibitors of gram-positive and gram-negative enoyl ACP reductases (FabI). The knowledge of the reported inhibitors can aid in the development of broad-spectrum antibacterials specifically targeting FabI enzymes from S. aureus, S. epidermidis, B. anthracis, B. cereus, E. coli, P. aeruginosa, P. falciparum and M. tuberculosis.


Asunto(s)
Antibacterianos/farmacología , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Compuestos Orgánicos/farmacología , Secuencia de Aminoácidos , Animales , Antibacterianos/química , Bacterias/enzimología , Línea Celular Tumoral , Enoil-ACP Reductasa (NADH)/química , Inhibidores Enzimáticos/química , Acido Graso Sintasa Tipo II/química , Humanos , Compuestos Orgánicos/química
6.
Nat Commun ; 11(1): 1727, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32265440

RESUMEN

Carbon-carbon bond forming reactions are essential transformations in natural product biosynthesis. During de novo fatty acid and polyketide biosynthesis, ß-ketoacyl-acyl carrier protein (ACP) synthases (KS), catalyze this process via a decarboxylative Claisen-like condensation reaction. KSs must recognize multiple chemically distinct ACPs and choreograph a ping-pong mechanism, often in an iterative fashion. Here, we report crystal structures of substrate mimetic bearing ACPs in complex with the elongating KSs from Escherichia coli, FabF and FabB, in order to better understand the stereochemical features governing substrate discrimination by KSs. Complemented by molecular dynamics (MD) simulations and mutagenesis studies, these structures reveal conformational states accessed during KS catalysis. These data taken together support a gating mechanism that regulates acyl-ACP binding and substrate delivery to the KS active site. Two active site loops undergo large conformational excursions during this dynamic gating mechanism and are likely evolutionarily conserved features in elongating KSs.


Asunto(s)
3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/química , Acetiltransferasas/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Acido Graso Sintasa Tipo II/química , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/aislamiento & purificación , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/metabolismo , Acetiltransferasas/metabolismo , Secuencia de Aminoácidos/genética , Sitios de Unión/genética , Catálisis , Dominio Catalítico/genética , Cristalografía por Rayos X , Escherichia coli/enzimología , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Acido Graso Sintasa Tipo II/metabolismo , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Simulación de Dinámica Molecular , Mutagénesis , Mutación , Conformación Proteica , Proteínas Recombinantes
7.
Protein Sci ; 29(5): 1242-1249, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32105377

RESUMEN

Urea amidolyase (UA), a bifunctional enzyme that is widely distributed in bacteria, fungi, algae, and plants, plays a pivotal role in the recycling of nitrogen in the biosphere. Its substrate urea is ultimately converted to ammonium, via successive catalysis at the C-terminal urea carboxylase (UC) domain and followed by the N-terminal allophanate hydrolyse (AH) domain. Although our previous studies have shown that Kluyveromyces lactis UA (KlUA) functions efficiently as a homodimer, the architecture of the full-length enzyme remains unresolved. Thus how the biotin carboxyl carrier protein (BCCP) domain is transferred within the UC domain remains unclear. Here we report the structures of full-length KlUA in its homodimer form in three different functional states by negatively-stained single-particle electron microscopy. We report here that the ADP-bound structure with or without urea shows two possible locations of BCCP with preferred asymmetry, and that when BCCP is attached to the carboxyl transferase domain of one monomer, it is attached to the biotin carboxylase domain in the second domain. Based on this observation, we propose a BCCP-swinging model for biotin-dependent carboxylation mechanism of this enzyme.


Asunto(s)
Ligasas de Carbono-Nitrógeno/metabolismo , Imagen Individual de Molécula , Acetil-CoA Carboxilasa/química , Acetil-CoA Carboxilasa/metabolismo , Biocatálisis , Ligasas de Carbono-Nitrógeno/química , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/metabolismo , Humanos , Conformación Proteica
8.
Biochem Biophys Res Commun ; 516(4): 1183-1189, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31296387

RESUMEN

Acyl carrier protein (ACP) is highly conserved across taxa and plays key roles in the fatty acid synthesis system by mediating acyl group delivery and shuttling. Here, we compared the structural and dynamic features of human type Ι ACP (hACP) and Escherichia coli type II ACP (EcACP). Analysis of chemical shift perturbations upon octanoyl group attachment showed perturbations in hACP only near acyl-group attachment sites, whereas EcACP showed the perturbation at residues in the hydrophobic cavity. This difference confirmed that hACP does not sequester the acyl chain in the hydrophobic cavity, which is blocked by hydrophobic triad residues (L34, L39, and V64). Moreover, hACP showed more flexible backbone dynamics than EcACP, especially in the front of α1α2 loop. We further investigated the interactions of hACP with Streptomyces coelicolor ACP synthase (ScAcpS), which is used to convert apo mammalian ACP to the holo form. Similar to protein-protein interface (PPI) found in hACP-hAcpS crystal structure, docking simulation and binding affinity measurements showed that the hydrophobic residues in universal recognition helix II of hACP contribute mainly to ScAcpS binding with binding affinity of 9.2 ±â€¯9.1 × 104 M. In contrast, interaction found in EcACP-EcAcpS crystal structure is dominated by electrostatic interactions. These results suggest that ScAcpS has relatively relaxed substrate specificity and a similar charge distribution to hAcpS. These fundamental differences of the charge distribution in hAcpS, ScAcpS and EcAcpS largely affect the interaction with hACP. These findings can provide a useful resource for development of novel antibiotics inhibiting PPI in bacterial FAS proteins with specificity.


Asunto(s)
Proteína Transportadora de Acilo/metabolismo , Proteínas Bacterianas/metabolismo , Escherichia coli/metabolismo , Ácidos Grasos/metabolismo , Streptomyces coelicolor/metabolismo , Proteína Transportadora de Acilo/química , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Escherichia coli/química , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Conformación Proteica , Mapas de Interacción de Proteínas , Alineación de Secuencia , Streptomyces coelicolor/química
9.
PLoS One ; 14(7): e0219435, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31291335

RESUMEN

Carrier proteins are four-helix bundles that covalently hold metabolites and secondary metabolites, such as fatty acids, polyketides and non-ribosomal peptides. These proteins mediate the production of many pharmaceutically important compounds including antibiotics and anticancer agents. Acyl carrier proteins (ACPs) can be found as part of a multi-domain polypeptide (Type I ACPs), or as part of a multiprotein complex (Type II). Here, the main focus is on ACP2 and ACP3, domains from the type I trans-AT polyketide synthase MmpA, which is a core component of the biosynthetic pathway of the antibiotic mupirocin. During molecular dynamics simulations of their apo, holo and acyl forms ACP2 and ACP3 both form a substrate-binding surface-groove. The substrates bound to this surface-groove have polar groups on their acyl chain exposed and forming hydrogen bonds with the solvent. Bulky hydrophobic residues in the GXDS motif common to all ACPs, and similar residues on helix III, appear to prohibit the formation of a deep tunnel in type I ACPs and type II ACPs from polyketide synthases. In contrast, the equivalent positions in ACPs from type II fatty acid synthases, which do form a deep solvent-excluded substrate-binding tunnel, have the small residue alanine. During simulation, ACP3 with mutations I61A L36A W44L forms a deep tunnel that can fully bury a saturated substrate in the core of the ACP, in contrast to the surface groove of the wild type ACP3. Similarly, in the ACP from E. coli fatty acid synthase, a type II ACP, mutations can change ligand binding from being inside a deep tunnel to being in a surface groove, thus demonstrating how changing a few residues can modify the possibilities for ligand binding.


Asunto(s)
Proteína Transportadora de Acilo/química , Complejos Multiproteicos/química , Péptidos/química , Sintasas Poliquetidas/química , Acinetobacter baumannii/química , Acinetobacter baumannii/genética , Proteína Transportadora de Acilo/genética , Proteína Transportadora de Acilo/metabolismo , Secuencias de Aminoácidos/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Vías Biosintéticas/genética , Secuestro de Carbono/genética , Escherichia coli/genética , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/genética , Acido Graso Sintasa Tipo II/metabolismo , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Simulación de Dinámica Molecular , Complejos Multiproteicos/genética , Mupirocina/biosíntesis , Mupirocina/metabolismo , Péptidos/genética , Mutación Puntual/genética , Sintasas Poliquetidas/genética , Unión Proteica
10.
Nat Chem Biol ; 15(7): 669-671, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31209348

RESUMEN

Fatty acid synthases are dynamic ensembles of enzymes that can biosynthesize long hydrocarbon chains efficiently. Here we visualize the interaction between the Escherichia coli acyl carrier protein (AcpP) and ß-ketoacyl-ACP-synthase I (FabB) using X-ray crystallography, NMR, and molecular dynamics simulations. We leveraged this structural information to alter lipid profiles in vivo and provide a molecular basis for how protein-protein interactions can regulate the fatty acid profile in E. coli.


Asunto(s)
3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/metabolismo , Proteína Transportadora de Acilo/metabolismo , Proteínas de Escherichia coli/metabolismo , Acido Graso Sintasa Tipo II/metabolismo , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/química , Proteína Transportadora de Acilo/química , Cristalografía por Rayos X , Escherichia coli/química , Escherichia coli/enzimología , Proteínas de Escherichia coli/química , Acido Graso Sintasa Tipo II/química , Modelos Moleculares , Unión Proteica
11.
Proc Natl Acad Sci U S A ; 116(14): 6775-6783, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30872475

RESUMEN

Fatty acid biosynthesis in α- and γ-proteobacteria requires two functionally distinct dehydratases, FabA and FabZ. Here, mechanistic cross-linking facilitates the structural characterization of a stable hexameric complex of six Escherichia coli FabZ dehydratase subunits with six AcpP acyl carrier proteins. The crystal structure sheds light on the divergent substrate selectivity of FabA and FabZ by revealing distinct architectures of the binding pocket. Molecular dynamics simulations demonstrate differential biasing of substrate orientations and conformations within the active sites of FabA and FabZ such that FabZ is preorganized to catalyze only dehydration, while FabA is primed for both dehydration and isomerization.


Asunto(s)
Proteína Transportadora de Acilo/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Acido Graso Sintasa Tipo II/química , Ácidos Grasos/química , Hidroliasas/química , Simulación de Dinámica Molecular , Complejos Multienzimáticos/química , Proteína Transportadora de Acilo/genética , Proteína Transportadora de Acilo/metabolismo , Catálisis , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Acido Graso Sintasa Tipo II/genética , Acido Graso Sintasa Tipo II/metabolismo , Ácidos Grasos/biosíntesis , Ácidos Grasos/genética , Hidroliasas/genética , Hidroliasas/metabolismo , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo
12.
Chembiochem ; 20(18): 2298-2321, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-30908841

RESUMEN

De novo biosynthesis of fatty acids is an iterative process requiring strict regulation of the lengths of the produced fatty acids. In this review, we focus on the factors determining chain lengths in fatty acid biosynthesis. In a nutshell, the process of chain-length regulation can be understood as the output of a chain-elongating C-C bond forming reaction competing with a terminating fatty acid release function. At the end of each cycle in the iterative process, the synthesizing enzymes need to "decide" whether the growing chain is to be elongated through another cycle or released as the "mature" fatty acid. Recent research has shed light on the factors determining fatty acid chain length and has also achieved control over chain length for the production of the technologically interesting short-chain (C4 -C8 ) and medium-chain (C10 -C14 ) fatty acids.


Asunto(s)
Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo I/química , Ácidos Grasos/biosíntesis , Proteína Transportadora de Acilo/química , Proteína Transportadora de Acilo/genética , Animales , Bacterias/enzimología , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo II/genética , Ácidos Grasos/química , Humanos , Estructura Molecular , Plantas/enzimología , Dominios Proteicos , Ingeniería de Proteínas , Saccharomyces cerevisiae/enzimología
13.
Eur J Med Chem ; 166: 108-118, 2019 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-30685534

RESUMEN

Thirty-six N-arylsulfonyl-3-substituted indoles were designed and synthesized by combining the N-arylsulfonylindoles with aminoguanidine, semicarbazide, and thiosemicarbazide, respectively. Their antibacterial activities were screened, and cytotoxic activities were evaluated. The results showed that aminoguanidines (6) exhibited much better antibacterial activity than semicarbazides (7) and thiosemicarbazides (8). Most compounds in series 6 showed potent inhibitory activity against the tested bacterial strains, including multidrug-resistant strains, with MIC values in the range of 1.08-23.46 µM. The cytotoxic activity of the compounds 6c, 6d, 6h, 6j, 6k and 6l was assessed in two human cancer cell lines A590 and SGC7901, and one human normal cell line HEK 293T. The results indicated that compounds selected exhibited excellent activity against the tested cancer cells with IC50 values in the range of 1.51-15.12 µM suggesting the potential of them as new antibacterial and anticancer agents. What's more, the results of resistance study revealed that resistance of the tested bacteria toward 6d is not easily developed. Molecular docking studies revealed that the aminoguanidine and arylsulfonylindole moieties played a significant role in binding the target site of E. coli FabH-CoA receptor.


Asunto(s)
Guanidinas/química , Indoles/síntesis química , Indoles/farmacología , Simulación del Acoplamiento Molecular , Semicarbacidas/química , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa , Acetiltransferasas/química , Acetiltransferasas/metabolismo , Antibacterianos/síntesis química , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Técnicas de Química Sintética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/metabolismo , Humanos , Indoles/química , Indoles/metabolismo , Pruebas de Sensibilidad Microbiana , Conformación Proteica , Relación Estructura-Actividad
14.
Biotechnol Lett ; 41(1): 181-191, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30498972

RESUMEN

OBJECTIVE: To enhance the thermostability and deregulate the hemin inhibition of 5-aminolevulinic acid (ALA) synthase from Rhodopseudomonas palustris (RP-ALAS) by a computer-aided rational design strategy. RESULTS: Eighteen RP-ALAS single variants were rationally designed and screened by measuring their residual activities upon heating. Among them, H29R and H15K exhibited a 2.3 °C and 6.0 °C higher melting temperature than wild-type, respectively. A 6.7-fold and 10.3-fold increase in specific activity after 1 h incubation at 37 °C was obtained for H29R (2.0 U/mg) and H15K (3.1 U/mg) compared to wild-type (0.3 U/mg). Additionally, higher residual activities in the presence of hemin were obtained for H29R and H15K (e.g., 64% and 76% at 10 µM hemin vs. 27% for wild-type). The ALA titer was increased by 6% and 22% in fermentation using Corynebacterium glutamicum ATCC 13032 expressing H29R and H15K, respectively. CONCLUSION: H29R and H15K showed high thermostability, reduced hemin inhibition and slightly high activity, indicating that these two variants are good candidates for bioproduction of ALA.


Asunto(s)
Sustitución de Aminoácidos , Proteínas Bacterianas/química , Simulación por Computador , Acido Graso Sintasa Tipo II/química , Hemina/química , Rhodopseudomonas/enzimología , Análisis de Secuencia de Proteína , Proteínas Bacterianas/genética , Estabilidad de Enzimas/genética , Acido Graso Sintasa Tipo II/genética , Calor , Rhodopseudomonas/genética
15.
Biochem Biophys Res Commun ; 505(1): 208-214, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30243724

RESUMEN

Bacterial fatty acid synthesis (FAS) has been extensively studied as a potential target of antimicrobials. In FAS, FabD mediates transacylation of the malonyl group from malonyl-CoA to acyl-carrier protein (ACP). The mounting threat of nosocomial infection by multidrug-resistant Acinetobacter baumannii warrants a deeper understanding of its essential cellular mechanisms, which could lead to effective control of this highly competent pathogen. The molecular mechanisms involved in A. baumannii FAS are poorly understood, and recent research has suggested that Pseudomonas aeruginosa, a closely related nosocomial pathogen of A. baumannii, utilizes FAS to produce virulence factors. In this study, we solved the crystal structure of A. baumannii FabD (AbFabD) to provide a platform for the development of new antibacterial agents. Analysis of the structure of AbFabD confirmed the presence of highly conserved active site residues among bacterial homologs. Binding constants between AbFabD variants and A. baumannii ACP (AbACP) revealed critical conserved residues Lys195 and Lys200 involved in AbACP binding. Computational docking of a potential inhibitor, trifluoperazine, revealed a unique inhibitor-binding pocket near the substrate-binding site. The structural study presented herein will be useful for the structure-based design of potent AbFabD inhibitors.


Asunto(s)
Acinetobacter baumannii/genética , S-Maloniltransferasa de la Proteína Transportadora de Grupos Acilo/genética , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana Múltiple/genética , Acido Graso Sintasa Tipo II/genética , Acinetobacter baumannii/enzimología , S-Maloniltransferasa de la Proteína Transportadora de Grupos Acilo/química , S-Maloniltransferasa de la Proteína Transportadora de Grupos Acilo/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión/genética , Cristalografía por Rayos X , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/metabolismo , Modelos Moleculares , Mutación , Dominios Proteicos , Homología de Secuencia de Aminoácido
16.
Nat Prod Rep ; 35(10): 1029-1045, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30046786

RESUMEN

Covering: up to April 5, 2018 Metabolites from type II fatty acid synthase (FAS) and polyketide synthase (PKS) pathways differ broadly in their identities and functional roles. The former are considered primary metabolites that are linear hydrocarbon acids, while the latter are complex aromatic or polyunsaturated secondary metabolites. Though the study of bacterial FAS has benefitted from decades of biochemical and structural investigations, type II PKSs have remained less understood. Here we review the recent approaches to understanding the protein-protein and protein-substrate interactions in these pathways, with an emphasis on recent chemical biology and structural applications. New approaches to the study of FAS have highlighted the critical role of the acyl carrier protein (ACP) with regard to how it stabilizes intermediates through sequestration and selectively delivers cargo to successive enzymes within these iterative pathways, utilizing protein-protein interactions to guide and organize enzymatic timing and specificity. Recent tools that have shown promise in FAS elucidation should find new approaches to studying type II PKS systems in the coming years.


Asunto(s)
Acido Graso Sintasa Tipo II/metabolismo , Sintasas Poliquetidas/metabolismo , Mapas de Interacción de Proteínas , Cristalografía por Rayos X , Ciclización , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/genética , Helicobacter pylori/metabolismo , Mutagénesis , Resonancia Magnética Nuclear Biomolecular , Sintasas Poliquetidas/química , Sintasas Poliquetidas/genética , Proteínas/análisis , Proteínas/química , Proteínas/genética , Especificidad por Sustrato
17.
ACS Synth Biol ; 7(5): 1179-1187, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29722970

RESUMEN

Medium-chain fatty acids (MCFAs) are key intermediates in the synthesis of medium-chain chemicals including α-olefins and dicarboxylic acids. In bacteria, microbial production of MCFAs is limited by the activity and product profile of fatty acyl-ACP thioesterases. Here, we engineer a heterologous bacterial medium-chain fatty acyl-ACP thioesterase for improved MCFA production in Escherichia coli. Electrostatically matching the interface between the heterologous medium-chain Acinetobacter baylyi fatty acyl-ACP thioesterase (AbTE) and the endogenous E. coli fatty acid ACP ( E. coli AcpP) by replacing small nonpolar amino acids on the AbTE surface for positively charged ones increased secreted MCFA titers more than 3-fold. Nuclear magnetic resonance titration of E. coli 15N-octanoyl-AcpP with a single AbTE point mutant and the best double mutant showed a progressive and significant increase in the number of interactions when compared to AbTE wildtype. The best AbTE mutant produced 131 mg/L of MCFAs, with MCFAs being 80% of all secreted fatty acid chain lengths after 72 h. To enable the future screening of larger numbers of AbTE variants to further improve MCFA titers, we show that a previously developed G-protein coupled receptor (GPCR)-based MCFA sensor differentially detects MCFAs secreted by E. coli expressing different AbTE variants. This work demonstrates that engineering the interface of heterologous enzymes to better couple with endogenous host proteins is a useful strategy to increase the titers of microbially produced chemicals. Further, this work shows that GPCR-based sensors are producer microbe agnostic and can detect chemicals directly in the producer microbe supernatant, setting the stage for the sensor-guided engineering of MCFA producing microbes.


Asunto(s)
Proteína Transportadora de Acilo/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Acido Graso Sintasa Tipo II/metabolismo , Ácidos Grasos/metabolismo , Ingeniería de Proteínas/métodos , Tioléster Hidrolasas/genética , Acinetobacter/enzimología , Acinetobacter/genética , Proteína Transportadora de Acilo/química , Proteína Transportadora de Acilo/genética , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/genética , Ácidos Grasos/genética , Espectroscopía de Resonancia Magnética , Microorganismos Modificados Genéticamente , Simulación del Acoplamiento Molecular , Mutación , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tioléster Hidrolasas/metabolismo
18.
Protein Sci ; 27(5): 969-975, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29520922

RESUMEN

FabA and FabZ are the two dehydratase enzymes in Escherichia coli that catalyze the dehydration of acyl intermediates in the biosynthesis of fatty acids. Both enzymes form obligate dimers in which the active site contains key amino acids from both subunits. While FabA is a soluble protein that has been relatively straightforward to express and to purify from cultured E. coli, FabZ has shown to be mostly insoluble and only partially active. In an effort to increase the solubility and activity of both dehydratases, we made constructs consisting of two identical subunits of FabA or FabZ fused with a naturally occurring peptide linker, so as to force their dimerization. The fused dimer of FabZ (FabZ-FabZ) was expressed as a soluble enzyme with an ninefold higher activity in vitro than the unfused FabZ. This construct exemplifies a strategy for the improvement of enzymes from the fatty acid biosynthesis pathways, many of which function as dimers, catalyzing critical steps for the production of fatty acids.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Acido Graso Sintasa Tipo II/metabolismo , Hidroliasas/metabolismo , Biocatálisis , Deshidratación , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/aislamiento & purificación , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/aislamiento & purificación , Ácidos Grasos/biosíntesis , Ácidos Grasos/química , Hidroliasas/química , Hidroliasas/aislamiento & purificación , Modelos Moleculares , Multimerización de Proteína , Solubilidad
19.
J Mol Biol ; 429(23): 3763-3775, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-29054754

RESUMEN

The Escherichia coli holo-(acyl carrier protein) synthase (ACPS) catalyzes the coenzyme A-dependent activation of apo-ACPP to generate holo-(acyl carrier protein) (holo-ACPP) in an early step of fatty acid biosynthesis. E. coli ACPS is sufficiently different from the human fatty acid synthase to justify the development of novel ACPS-targeting antibiotics. Models of E. coli ACPS in unliganded and holo-ACPP-bound forms solved by X-ray crystallography to 2.05and 4.10Å, respectively, revealed that ACPS bound three product holo-ACPP molecules to form a 3:3 hexamer. Solution NMR spectroscopy experiments validated the ACPS binding interface on holo-ACPP using chemical shift perturbations and by determining the relative orientation of holo-ACPP to ACPS by fitting residual dipolar couplings. The binding interface is organized to arrange contacts between positively charged ACPS residues and the holo-ACPP phosphopantetheine moiety, indicating product contains more stabilizing interactions than expected in the enzyme:substrate complex. Indeed, holo-ACPP bound the enzyme with greater affinity than the substrate, apo-ACPP, and with negative cooperativity. The first equivalent of holo-ACPP bound with a KD=62±13nM, followed by the binding of two more equivalents of holo-ACPP with KD=1.2±0.2µM. Cooperativity was not observed for apo-ACPP which bound with KD=2.4±0.1µM. Strong product binding and high levels of holo-ACPP in the cell identify a potential regulatory role of ACPS in fatty acid biosynthesis.


Asunto(s)
Proteína Transportadora de Acilo/química , Proteína Transportadora de Acilo/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/metabolismo , Transferasas/química , Transferasas/metabolismo , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/crecimiento & desarrollo , Modelos Moleculares , Unión Proteica , Conformación Proteica
20.
Acta Crystallogr D Struct Biol ; 73(Pt 9): 757-766, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28876239

RESUMEN

An alternative rational approach to improve protein crystals by using single-site mutation of surface residues is proposed based on the results of a statistical analysis using a compiled data set of 918 independent crystal structures, thereby reflecting not only the entropic effect but also other effects upon protein crystallization. This analysis reveals a clear difference in the crystal-packing propensity of amino acids depending on the secondary-structural class. To verify this result, a systematic crystallization experiment was performed with the biotin carboxyl carrier protein from Pyrococcus horikoshii OT3 (PhBCCP). Six single-site mutations were examined: Ala138 on the surface of a ß-sheet was mutated to Ile, Tyr, Arg, Gln, Val and Lys. In agreement with prediction, it was observed that the two mutants (A138I and A138Y) harbouring the residues with the highest crystal-packing propensities for ß-sheet at position 138 provided better crystallization scores relative to those of other constructs, including the wild type, and that the crystal-packing propensity for ß-sheet provided the best correlation with the ratio of obtaining crystals. Two new crystal forms of these mutants were obtained that diffracted to high resolution, generating novel packing interfaces with the mutated residues (Ile/Tyr). The mutations introduced did not affect the overall structures, indicating that a ß-sheet can accommodate a successful mutation if it is carefully selected so as to avoid intramolecular steric hindrance. A significant negative correlation between the ratio of obtaining amorphous precipitate and the crystal-packing propensity was also found.


Asunto(s)
Acetil-CoA Carboxilasa/química , Proteínas Arqueales/química , Pyrococcus horikoshii/química , Acetil-CoA Carboxilasa/genética , Aminoácidos/química , Aminoácidos/genética , Proteínas Arqueales/genética , Cristalografía por Rayos X , Acido Graso Sintasa Tipo II/química , Acido Graso Sintasa Tipo II/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Conformación Proteica , Estructura Secundaria de Proteína , Pyrococcus horikoshii/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA