Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(23): e2405555121, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38805268

RESUMEN

The dimeric nuclear factor kappa B (NF-κB) transcription factors (TFs) regulate gene expression by binding to a variety of κB DNA elements with conserved G:C-rich flanking sequences enclosing a degenerate central region. Toward defining mechanistic principles of affinity regulated by degeneracy, we observed an unusual dependence of the affinity of RelA on the identity of the central base pair, which appears to be noncontacted in the complex crystal structures. The affinity of κB sites with A or T at the central position is ~10-fold higher than with G or C. The crystal structures of neither the complexes nor the free κB DNAs could explain the differences in affinity. Interestingly, differential dynamics of several residues were revealed in molecular dynamics simulation studies, where simulation replicates totaling 148 µs were performed on NF-κB:DNA complexes and free κB DNAs. Notably, Arg187 and Arg124 exhibited selectivity in transient interactions that orchestrated a complex interplay among several DNA-interacting residues in the central region. Binding and simulation studies with mutants supported these observations of transient interactions dictating specificity. In combination with published reports, this work provides insights into the nuanced mechanisms governing the discriminatory binding of NF-κB family TFs to κB DNA elements and sheds light on cancer pathogenesis of cRel, a close homolog of RelA.


Asunto(s)
ADN , Simulación de Dinámica Molecular , FN-kappa B , Unión Proteica , ADN/metabolismo , Humanos , FN-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Sitios de Unión , Cristalografía por Rayos X
2.
Nat Prod Rep ; 41(1): 113-147, 2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-37929638

RESUMEN

Covering: 2009 up to August 2023Prenyltransferases (PTs) are involved in the primary and the secondary metabolism of plants, bacteria, and fungi, and they are key enzymes in the biosynthesis of many clinically relevant natural products (NPs). The continued biochemical and structural characterization of the soluble dimethylallyl tryptophan synthase (DMATS) PTs over the past two decades have revealed the significant promise that these enzymes hold as biocatalysts for the chemoenzymatic synthesis of novel drug leads. This is a comprehensive review of DMATSs describing the structure-function relationships that have shaped the mechanistic underpinnings of these enzymes, as well as the application of this knowledge to the engineering of DMATSs. We summarize the key findings and lessons learned from these studies over the past 14 years (2009-2023). In addition, we identify current gaps in our understanding of these fascinating enzymes.


Asunto(s)
Dimetilaliltranstransferasa , Dimetilaliltranstransferasa/química , Prenilación , Hongos/metabolismo
3.
FASEB J ; 37(5): e22914, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37043381

RESUMEN

Thiol isomerases, including PDI, ERp57, ERp5, and ERp72, play important and distinct roles in cancer progression, cancer cell signaling, and metastasis. We recently discovered that zafirlukast, an FDA-approved medication for asthma, is a pan-thiol isomerase inhibitor. Zafirlukast inhibited the growth of multiple cancer cell lines with an IC50 in the low micromolar range, while also inhibiting cellular thiol isomerase activity, EGFR activation, and downstream phosphorylation of Gab1. Zafirlukast also blocked the procoagulant activity of OVCAR8 cells by inhibiting tissue factor-dependent Factor Xa generation. In an ovarian cancer xenograft model, statistically significant differences in tumor size between control vs treated groups were observed by Day 18. Zafirlukast also significantly reduced the number and size of metastatic tumors found within the lungs of the mock-treated controls. When added to a chemotherapeutic regimen, zafirlukast significantly reduced growth, by 38% compared with the mice receiving only the chemotherapeutic treatment, and by 83% over untreated controls. Finally, we conducted a pilot clinical trial in women with tumor marker-only (CA-125) relapsed ovarian cancer, where the rate of rise of CA-125 was significantly reduced following treatment with zafirlukast, while no severe adverse events were reported. Thiol isomerase inhibition with zafirlukast represents a novel, well-tolerated therapeutic in the treatment of ovarian cancer.


Asunto(s)
Plaquetas , Neoplasias Ováricas , Animales , Femenino , Humanos , Ratones , Plaquetas/metabolismo , Indoles , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Fenilcarbamatos/metabolismo , Compuestos de Sulfhidrilo/metabolismo
4.
Biochemistry ; 62(1): 109-117, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36525630

RESUMEN

The Gram-positive pathogen Staphylococcus aureus is a leading cause of antimicrobial resistance related deaths worldwide. Like many pathogens with multidrug-resistant strains, S. aureus contains enzymes that confer resistance through antibiotic modification(s). One such enzyme present in S. aureus is FosB, a Mn2+-dependent l-cysteine or bacillithiol (BSH) transferase that inactivates the antibiotic fosfomycin. fosB gene knockout experiments show that the minimum inhibitory concentration (MIC) of fosfomycin is significantly reduced when the FosB enzyme is not present. This suggests that inhibition of FosB could be an effective method to restore fosfomycin activity. We used high-throughput in silico-based screening to identify small-molecule analogues of fosfomycin that inhibited thiol transferase activity. Phosphonoformate (PPF) was a top hit from our approach. Herein, we have characterized PPF as a competitive inhibitor of FosB from S. aureus (FosBSa) and Bacillus cereus (FosBBc). In addition, we have determined a crystal structure of FosBBc with PPF bound in the active site. Our results will be useful for future structure-based development of FosB inhibitors that can be delivered in combination with fosfomycin in order to increase the efficacy of this antibiotic.


Asunto(s)
Fosfomicina , Antibacterianos/química , Foscarnet/metabolismo , Foscarnet/farmacología , Fosfomicina/química , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus/metabolismo , Transferasas/metabolismo , Farmacorresistencia Bacteriana , Proteínas Bacterianas/metabolismo
5.
Biochemistry ; 62(3): 710-721, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36657084

RESUMEN

Over one and a half million people die of tuberculosis (TB) each year. Multidrug-resistant TB infections are especially dangerous, and new drugs are needed to combat them. The high cost and complexity of drug development make repositioning of drugs that are already in clinical use for other indications a potentially time- and money-saving avenue. In this study, we identified among existing drugs five compounds: azelastine, venlafaxine, chloroquine, mefloquine, and proguanil as inhibitors of acetyltransferase Eis from Mycobacterium tuberculosis, a causative agent of TB. Eis upregulation is a cause of clinically relevant resistance of TB to kanamycin, which is inactivated by Eis-catalyzed acetylation. Crystal structures of these drugs as well as chlorhexidine in complexes with Eis showed that these inhibitors were bound in the aminoglycoside binding cavity, consistent with their established modes of inhibition with respect to kanamycin. Among three additionally synthesized compounds, a proguanil analogue, designed based on the crystal structure of the Eis-proguanil complex, was 3-fold more potent than proguanil. The crystal structures of these compounds in complexes with Eis explained their inhibitory potencies. These initial efforts in rational drug repositioning can serve as a starting point in further development of Eis inhibitors.


Asunto(s)
Acetiltransferasas , Mycobacterium tuberculosis , Tuberculosis , Humanos , Acetiltransferasas/antagonistas & inhibidores , Antituberculosos/farmacología , Antituberculosos/química , Proteínas Bacterianas/antagonistas & inhibidores , Kanamicina/farmacología , Kanamicina/química , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Proguanil/metabolismo , Tuberculosis/tratamiento farmacológico
6.
Biochemistry ; 2020 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-33175491

RESUMEN

ETS family transcription factors control development of different cell types in humans, whereas deregulation of these proteins leads to severe developmental syndromes and cancers. One of a few members of the ETS family that are known to act solely as repressors, ERF, is required for normal osteogenesis and hematopoiesis. Another important function of ERF is acting as a tumor suppressor by antagonizing oncogenic fusions involving other ETS family factors. The structure of ERF and the DNA binding properties specific to this protein have not been elucidated. In this study, we determined two crystal structures of the complexes of the DNA binding domain of ERF with DNA. In one, ERF is in a distinct dimeric form, with Cys72 in a reduced state. In the other, two dimers of ERF are assembled into a tetramer that is additionally locked by two Cys72-Cys72 disulfide bonds across the dimers. In the tetramer, the ERF molecules are bound to a pseudocontinuous DNA on the same DNA face at two GGAA binding sites on opposite strands. Sedimentation velocity analysis showed that this tetrameric assembly forms on continuous DNA containing such tandem sites spaced by 7 bp. Our bioinformatic analysis of three previously reported sets of ERF binding loci across entire genomes showed that these loci were enriched in such 7 bp spaced tandem sites. Taken together, these results strongly suggest that the observed tetrameric assembly is a functional state of ERF in the human cell.

7.
Nat Chem Biol ; 14(5): 428-430, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29556104

RESUMEN

Interrupted adenylation domains are enigmatic fusions, in which one enzyme is inserted into another to form a highly unusual bifunctional enzyme. We present the first crystal structure of an interrupted adenylation domain that reveals a unique embedded methyltransferase. The structure and functional data provide insight into how these enzymes N-methylate amino acid precursors en route to nonribosomal peptides.


Asunto(s)
Aminoácidos/química , Enzimas/química , Metilación , Péptidos/química , Adenosina Monofosfato/química , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Escherichia coli/metabolismo , Iminas/química , Cinética , Péptido Sintasas/química , Dominios Proteicos , Especificidad por Sustrato , Factores de Tiempo
8.
Angew Chem Int Ed Engl ; 59(2): 826-832, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31702856

RESUMEN

MtmOIV and MtmW catalyze the final two reactions in the mithramycin (MTM) biosynthetic pathway, the Baeyer-Villiger opening of the fourth ring of premithramycin B (PMB), creating the C3 pentyl side chain, strictly followed by reduction of the distal keto group on the new side chain. Unexpectedly this results in a C2 stereoisomer of mithramycin, iso-mithramycin (iso-MTM). Iso-MTM undergoes a non-enzymatic isomerization to MTM catalyzed by Mg2+ ions. Crystal structures of MtmW and its complexes with co-substrate NADPH and PEG, suggest a catalytic mechanism of MtmW. The structures also show that a tetrameric assembly of this enzyme strikingly resembles the ring-shaped ß subunit of a vertebrate ion channel. We show that MtmW and MtmOIV form a complex in the presence of PMB and NADPH, presumably to hand over the unstable MtmOIV product to MtmW, yielding iso-MTM, as a potential self-resistance mechanism against MTM toxicity.


Asunto(s)
Productos Biológicos/metabolismo , Plicamicina/biosíntesis , Catálisis
9.
Molecules ; 25(1)2019 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-31878304

RESUMEN

The fact that the number of people with Alzheimer's disease is increasing, combined with the limited availability of drugs for its treatment, emphasize the need for the development of novel effective therapeutics for treating this brain disorder. Herein, we focus on generating 12 chalcone-donepezil hybrids, with the goal of simultaneously targeting amyloid-ß (Aß) peptides as well as cholinesterases (i.e., acetylcholinesterase (AChE) and butyrylcholinesterase (BChE)). We present the design, synthesis, and biochemical evaluation of these two series of novel 1,3-chalcone-donepezil (15a-15f) or 1,4-chalcone-donepezil (16a-16f) hybrids. We evaluate the relationship between their structures and their ability to inhibit AChE/BChE activity as well as their ability to bind Aß peptides. We show that several of these novel chalcone-donepezil hybrids can successfully inhibit AChE/BChE as well as the assembly of N-biotinylated Aß(1-42) oligomers. We also demonstrate that the Aß binding site of these hybrids differs from that of Pittsburgh Compound B (PIB).


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Chalconas/farmacología , Inhibidores de la Colinesterasa/farmacología , Donepezilo/farmacología , Acetilcolinesterasa/metabolismo , Péptidos beta-Amiloides/efectos de los fármacos , Compuestos de Anilina/química , Butirilcolinesterasa/metabolismo , Chalconas/síntesis química , Chalconas/química , Donepezilo/síntesis química , Donepezilo/química , Humanos , Modelos Moleculares , Tiazoles/química , Tritio/metabolismo
10.
Biochemistry ; 57(14): 2084-2093, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29558114

RESUMEN

Bacterial primase DnaG is an essential nucleic acid polymerase that generates primers for replication of chromosomal DNA. The mechanism of DnaG remains unclear due to the paucity of structural information on DnaG in complexes with other replisome components. Here we report the first crystal structures of noncovalent DnaG-DNA complexes, obtained with the RNA polymerase domain of Mycobacterium tuberculosis DnaG and various DNA ligands. One structure, obtained with ds DNA, reveals interactions with DnaG as it slides on ds DNA and suggests how DnaG binds template for primer synthesis. In another structure, DNA in the active site of DnaG mimics the primer, providing insight into mechanisms for the nucleotide transfer and DNA translocation. In conjunction with the recent cryo-EM structure of the bacteriophage T7 replisome, this study yields a model for primer elongation and hand-off to DNA polymerase.


Asunto(s)
Proteínas Bacterianas/química , ADN Primasa/química , ADN Bacteriano/química , Modelos Moleculares , Mycobacterium tuberculosis/enzimología , Proteínas Bacterianas/metabolismo , ADN Primasa/metabolismo , ADN Bacteriano/biosíntesis , Modelos Químicos , Dominios Proteicos
11.
Biochemistry ; 57(5): 781-790, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29345920

RESUMEN

Bacterial nucleoid-associated proteins (NAPs) are critical to genome integrity and chromosome maintenance. Post-translational modifications of bacterial NAPs appear to function similarly to their better studied mammalian counterparts. The histone-like NAP HupB from Mycobacterium tuberculosis (Mtb) was previously observed to be acetylated by the acetyltransferase Eis, leading to genome reorganization. We report biochemical and structural aspects of acetylation of HupB by Eis. We also found that the SirT-family NAD+-dependent deacetylase Rv1151c from Mtb deacetylated HupB in vitro and characterized the deacetylation kinetics. We propose that activities of Eis and Rv1151c could regulate the acetylation status of HupB to remodel the mycobacterial chromosome in response to environmental changes.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Mycobacterium tuberculosis/metabolismo , Acetilación , Acetiltransferasas/antagonistas & inhibidores , Acetiltransferasas/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Clonación Molecular , Cristalografía por Rayos X , Farmacorresistencia Bacteriana Múltiple/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/fisiología , Histona Desacetilasas/genética , Histonas/genética , Cinética , Lisina/química , Modelos Moleculares , Mycobacterium tuberculosis/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Espectrometría de Masas en Tándem
12.
Nucleic Acids Res ; 44(18): 8990-9004, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27587584

RESUMEN

Transcription factors have been considered undruggable, but this paradigm has been recently challenged. DNA binding natural product mithramycin (MTM) is a potent antagonist of oncogenic transcription factor EWS-FLI1. Structural details of MTM recognition of DNA, including the FLI1 binding sequence GGA(A/T), are needed to understand how MTM interferes with EWS-FLI1. We report a crystal structure of an MTM analogue MTM SA-Trp bound to a DNA oligomer containing a site GGCC, and two structures of a novel analogue MTM SA-Phe in complex with DNA. MTM SA-Phe is bound to sites AGGG and GGGT on one DNA, and to AGGG and GGGA(T) (a FLI1 binding site) on the other, revealing how MTM recognizes different DNA sequences. Unexpectedly, at sub-micromolar concentrations MTMs stabilize FLI1-DNA complex on GGAA repeats, which are critical for the oncogenic function of EWS-FLI1. We also directly demonstrate by nuclear magnetic resonance formation of a ternary FLI1-DNA-MTM complex on a single GGAA FLI1/MTM binding site. These biochemical and structural data and a new FLI1-DNA structure suggest that MTM binds the minor groove and perturbs FLI1 bound nearby in the major groove. This ternary complex model may lead to development of novel MTM analogues that selectively target EWS-FLI1 or other oncogenic transcription factors, as anti-cancer therapeutics.


Asunto(s)
ADN/química , Plicamicina/química , Proteína Proto-Oncogénica c-fli-1/química , Secuencia de Bases , ADN/metabolismo , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/metabolismo , Plicamicina/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteína Proto-Oncogénica c-fli-1/metabolismo , Relación Estructura-Actividad
13.
Biochemistry ; 56(46): 6087-6097, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29112395

RESUMEN

Dimethylation of amino acids consists of an interesting and puzzling series of events that could be achieved, during nonribosomal peptide biosynthesis, either by a single adenylation (A) domain interrupted by a methyltransferase (M) domain or by the sequential action of two of such independent enzymes. Herein, to establish the method by which Nature N,S-dimethylates l-Cys, we studied its formation during thiochondrilline A biosynthesis by evaluating TioS(A3aM3SA3bT3) and TioN(AaMNAb). This study not only led to identification of the exact pathway followed in Nature by these two enzymes for N,S-dimethylation of l-Cys, but also revealed that a single interrupted A domain can N,N-dimethylate amino acids, a novel phenomenon in the nonribosomal peptide field. These findings offer important and useful insights for the development and engineering of novel interrupted A domain enzymes to serve, in the future, as tools for combinatorial biosynthesis.


Asunto(s)
Cisteína/metabolismo , Hidroxiquinolinas/metabolismo , Micromonosporaceae/enzimología , Micromonosporaceae/metabolismo , Oligopéptidos/metabolismo , Péptido Sintasas/metabolismo , Vías Biosintéticas , Metilación , Biosíntesis de Péptidos Independientes de Ácidos Nucleicos , Péptido Sintasas/química , Dominios Proteicos
14.
Org Biomol Chem ; 14(1): 74-84, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26548370

RESUMEN

Advanced prostate tumors usually metastasize to the lung, bone, and other vital tissues and are resistant to conventional therapy. Prostate apoptosis response-4 protein (Par-4) is a tumor suppressor that causes apoptosis in therapy-resistant prostate cancer cells by binding specifically to a receptor, Glucose-regulated protein-78 (GRP78), found only on the surface of cancer cells. 3-Arylquinolines or "arylquins" induce normal cells to release Par-4 from the intermediate filament protein, vimentin and promote Par-4 secretion that targets cancer cells in a paracrine manner. A structure-activity study identified arylquins that promote Par-4 secretion, and an evaluation of arylquin binding to the hERG potassium ion channel using a [(3)H]-dofetilide binding assay permitted the identification of structural features that separated this undesired activity from the desired Par-4 secretory activity. A binding study that relied on the natural fluorescence of arylquins and that used the purified rod domain of vimentin (residues 99-411) suggested that the mechanism behind Par-4 release involved arylquin binding to multiple sites in the rod domain.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Quinolonas/metabolismo , Quinolonas/farmacología , Vimentina/metabolismo , Sitios de Unión/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Estructura Molecular , Quinolonas/química , Estereoisomerismo , Relación Estructura-Actividad , Vimentina/química
15.
Proc Natl Acad Sci U S A ; 110(42): 16784-9, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043778

RESUMEN

Protein folding has been extensively studied, but many questions remain regarding the mechanism. Characterizing early unstable intermediates and the high-free-energy transition state (TS) will help answer some of these. Here, we use effects of denaturants (urea, guanidinium chloride) and temperature on folding and unfolding rate constants and the overall equilibrium constant as probes of surface area changes in protein folding. We interpret denaturant kinetic m-values and activation heat capacity changes for 13 proteins to determine amounts of hydrocarbon and amide surface buried in folding to and from TS, and for complete folding. Predicted accessible surface area changes for complete folding agree in most cases with structurally determined values. We find that TS is advanced (50-90% of overall surface burial) and that the surface buried is disproportionately amide, demonstrating extensive formation of secondary structure in early intermediates. Models of possible pre-TS intermediates with all elements of the native secondary structure, created for several of these proteins, bury less amide and hydrocarbon surface than predicted for TS. Therefore, we propose that TS generally has both the native secondary structure and sufficient organization of other regions of the backbone to nucleate subsequent (post-TS) formation of tertiary interactions. The approach developed here provides proof of concept for the use of denaturants and other solutes as probes of amount and composition of the surface buried in coupled folding and other large conformational changes in TS and intermediates in protein processes.


Asunto(s)
Modelos Químicos , Desnaturalización Proteica , Pliegue de Proteína , Proteínas/química
16.
Proc Natl Acad Sci U S A ; 110(28): 11320-5, 2013 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-23801751

RESUMEN

Functional assignment of enzymes encoded by the Mycobacterium tuberculosis genome is largely incomplete despite recent advances in genomics and bioinformatics. Here, we applied an activity-based metabolomic profiling method to assign function to a unique phosphatase, Rv1692. In contrast to its annotation as a nucleotide phosphatase, metabolomic profiling and kinetic characterization indicate that Rv1692 is a D,L-glycerol 3-phosphate phosphatase. Crystal structures of Rv1692 reveal a unique architecture, a fusion of a predicted haloacid dehalogenase fold with a previously unidentified GCN5-related N-acetyltransferase region. Although not directly involved in acetyl transfer, or regulation of enzymatic activity in vitro, this GCN5-related N-acetyltransferase region is critical for the solubility of the phosphatase. Structural and biochemical analysis shows that the active site features are adapted for recognition of small polyol phosphates, and not nucleotide substrates. Functional assignment and metabolomic studies of M. tuberculosis lacking rv1692 demonstrate that Rv1692 is the final enzyme involved in glycerophospholipid recycling/catabolism, a pathway not previously described in M. tuberculosis.


Asunto(s)
Glicerofosfolípidos/metabolismo , Mycobacterium tuberculosis/enzimología , Monoéster Fosfórico Hidrolasas/metabolismo , Dominio Catalítico , Modelos Moleculares , Monoéster Fosfórico Hidrolasas/química , Solubilidad
17.
Biochemistry ; 54(50): 7365-74, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26618620

RESUMEN

FLI1 (Friend leukemia integration 1) is a metazoan transcription factor that is upregulated in a number of cancers. In addition, rearrangements of the fli1 gene cause sarcomas, leukemias, and lymphomas. These rearrangements encode oncogenic transcription factors, in which the DNA binding domain (DBD or ETS domain) of FLI1 on the C-terminal side is fused to a part of an another protein on the N-terminal side. Such abnormal cancer cell-specific fusions retain the DNA binding properties of FLI1 and acquire non-native protein-protein or protein-nucleic acid interactions of the substituted region. As a result, these fusions trigger oncogenic transcriptional reprogramming of the host cell. Interactions of FLI1 fusions with other proteins and with itself play a critical role in the oncogenic regulatory functions, and they are currently under intense scrutiny, mechanistically and as potential novel anticancer drug targets. We report elusive crystal structures of the FLI1 DBD, alone and in complex with cognate DNA containing a GGAA recognition sequence. Both structures reveal a previously unrecognized dimer of this domain, consistent with its dimerization in solution. The homodimerization interface is helix-swapped and dominated by hydrophobic interactions, including those between two interlocking Phe362 residues. A mutation of Phe362 to an alanine disrupted the propensity of this domain to dimerize without perturbing its structure or the DNA binding function, consistent with the structural observations. We propose that FLI1 DBD dimerization plays a role in transcriptional activation and repression by FLI1 and its fusions at promoters containing multiple FLI1 binding sites.


Asunto(s)
ADN/metabolismo , Proteína Proto-Oncogénica c-fli-1/química , Proteína Proto-Oncogénica c-fli-1/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Homología de Secuencia de Aminoácido
18.
Biochemistry ; 54(15): 2481-9, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25587924

RESUMEN

More and more post-PKS tailoring enzymes are recognized as being multifunctional and codependent on other tailoring enzymes. One of the recently discovered intriguing examples is MtmC, a bifunctional TDP-4-keto-d-olivose ketoreductase-methyltransferase, which-in codependence with glycosyltransferase MtmGIV-is a key contributor to the biosynthesis of the critical trisaccharide chain of the antitumor antibiotic mithramycin (MTM), produced by Streptomyces argillaceus. We report crystal structures of three binary complexes of MtmC with its methylation cosubstrate SAM, its coproduct SAH, and a nucleotide TDP as well as crystal structures of two ternary complexes, MtmC-SAH-TDP-4-keto-d-olivose and MtmC-SAM-TDP, in the range of 2.2-2.7 Å resolution. The structures reveal general and sugar-specific recognition and catalytic structural features of MtmC. Depending on the catalytic function that is conducted by MtmC, it must bind either NADPH or SAM in the same cofactor binding pocket. A tyrosine residue (Tyr79) appears as a lid covering the sugar moiety of the substrate during the methyl transfer reaction. This residue swings out of the active site by ~180° in the absence of the substrate. This unique conformational change likely serves to release the methylated product and, possibly, to open the active site for binding the bulkier cosubstrate NADPH prior to the reduction reaction.


Asunto(s)
Proteínas Bacterianas/química , Metiltransferasas/química , Oxidorreductasas/química , Streptomyces/enzimología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , NADP/química , NADP/genética , NADP/metabolismo , Oxidación-Reducción , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Plicamicina/biosíntesis , Streptomyces/genética , Relación Estructura-Actividad
19.
Biochemistry ; 54(20): 3197-206, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25928210

RESUMEN

Proteins from the enhanced intracellular survival (Eis) family are versatile acetyltransferases that acetylate amines at multiple positions of several aminoglycosides (AGs). Their upregulation confers drug resistance. Homologues of Eis are present in diverse bacteria, including many pathogens. Eis from Mycobacterium tuberculosis (Eis_Mtb) has been well characterized. In this study, we explored the AG specificity and catalytic efficiency of the Eis family protein from Bacillus anthracis (Eis_Ban). Kinetic analysis of specificity and catalytic efficiency of acetylation of six AGs indicates that Eis_Ban displays significant differences from Eis_Mtb in both substrate binding and catalytic efficiency. The number of acetylated amines was also different for several AGs, indicating a distinct regiospecificity of Eis_Ban. Furthermore, most recently identified inhibitors of Eis_Mtb did not inhibit Eis_Ban, underscoring the differences between these two enzymes. To explain these differences, we determined an Eis_Ban crystal structure. The comparison of the crystal structures of Eis_Ban and Eis_Mtb demonstrates that critical residues lining their respective substrate binding pockets differ substantially, explaining their distinct specificities. Our results suggest that acetyltransferases of the Eis family evolved divergently to garner distinct specificities while conserving catalytic efficiency, possibly to counter distinct chemical challenges. The unique specificity features of these enzymes can be utilized as tools for developing AGs with novel modifications and help guide specific AG treatments to avoid Eis-mediated resistance.


Asunto(s)
Acetiltransferasas/química , Bacillus anthracis/enzimología , Proteínas Bacterianas/química , Acetilación , Acetiltransferasas/antagonistas & inhibidores , Acetiltransferasas/metabolismo , Secuencia de Aminoácidos , Antibacterianos/farmacología , Bacillus anthracis/efectos de los fármacos , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Farmacorresistencia Bacteriana , Concentración 50 Inhibidora , Cinética , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , Estructura Secundaria de Proteína
20.
J Struct Biol ; 192(3): 349-357, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26416533

RESUMEN

Pyoluteorin is an antifungal agent composed of a 4,5-dichlorinated pyrrole group linked to a resorcinol moiety. The pyoluteorin biosynthetic gene cluster in Pseudomonas fluorescens Pf-5 encodes the halogenase PltA, which has been previously demonstrated to perform both chlorinations in vitro. PltA selectively accepts as a substrate a pyrrole moiety covalently tethered to a nonribosomal peptide thiolation domain PltL (pyrrolyl-S-PltL) for FAD-dependent di-chlorination, yielding 4,5-dichloropyrrolyl-S-PltL. We report a 2.75 Å-resolution crystal structure of PltA in complex with FAD and chloride. PltA is a dimeric enzyme, containing a flavin-binding fold conserved in flavin-dependent halogenases and monooxygenases, and an additional unique helical region at the C-terminus. This C-terminal region blocks a putative substrate-binding cleft, suggesting that a conformational change involving repositioning of this region is necessary to allow binding of the pyrrolyl-S-PltL substrate for its dichlorination by PltA.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/ultraestructura , Cloro/química , Flavina-Adenina Dinucleótido/química , Halogenación/fisiología , Fenoles/química , Pirroles/química , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Oxidación-Reducción , Oxidorreductasas/química , Unión Proteica/fisiología , Pseudomonas fluorescens/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA