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.
Proc Natl Acad Sci U S A ; 120(29): e2304378120, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37428913

RESUMEN

ToxR, a Vibrio cholerae transmembrane one-component signal transduction factor, lies within a regulatory cascade that results in the expression of ToxT, toxin coregulated pilus, and cholera toxin. While ToxR has been extensively studied for its ability to activate or repress various genes in V. cholerae, here we present the crystal structures of the ToxR cytoplasmic domain bound to DNA at the toxT and ompU promoters. The structures confirm some predicted interactions, yet reveal other unexpected promoter interactions with implications for other potential regulatory roles for ToxR. We show that ToxR is a versatile virulence regulator that recognizes diverse and extensive, eukaryotic-like regulatory DNA sequences, that relies more on DNA structural elements than specific sequences for binding. Using this topological DNA recognition mechanism, ToxR can bind both in tandem and in a twofold inverted-repeat-driven manner. Its regulatory action is based on coordinated multiple binding to promoter regions near the transcription start site, which can remove the repressing H-NS proteins and prepares the DNA for optimal interaction with the RNA polymerase.


Asunto(s)
Vibrio cholerae , Vibrio cholerae/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/metabolismo , Virulencia , Proteínas Bacterianas/metabolismo , ADN/genética , ADN/metabolismo , Regulación Bacteriana de la Expresión Génica
2.
Nucleic Acids Res ; 51(3): 1458-1472, 2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36688326

RESUMEN

DNA replication is essential to all living organisms as it ensures the fidelity of genetic material for the next generation of dividing cells. One of the simplest replication initiation mechanisms is the rolling circle replication. In the streptococcal plasmid pMV158, which confers antibiotic resistance to tetracycline, replication initiation is catalysed by RepB protein. The RepB N-terminal domain or origin binding domain binds to the recognition sequence (bind locus) of the double-strand origin of replication and cleaves one DNA strand at a specific site within the nic locus. Using biochemical and crystallographic analyses, here we show how the origin binding domain recognises and binds to the bind locus using structural elements removed from the active site, namely the recognition α helix, and a ß-strand that organises upon binding. A new hexameric structure of full-length RepB that highlights the great flexibility of this protein is presented, which could account for its ability to perform different tasks, namely bind to two distinct loci and cleave one strand of DNA at the plasmid origin.


Asunto(s)
Replicación del ADN , Plásmidos , Streptococcus , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , ADN Bacteriano/metabolismo , Origen de Réplica , Streptococcus/genética
3.
Antiviral Res ; 208: 105458, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36336176

RESUMEN

Severe acute respiratory syndrome coronaviruses 1 and 2 (SARS-CoV-1 and SARS-CoV-2) pose a threat to global public health. The 3C-like main protease (Mpro), which presents structural similarity with the active site domain of enterovirus 3C protease, is one of the best-characterized drug targets of these viruses. Here we studied the antiviral activity of the orally bioavailable enterovirus protease inhibitor AG7404 against SARS-CoV-1 and SARS-CoV-2 from a structural, biochemical, and cellular perspective, comparing it with the related molecule rupintrivir (AG7800). Crystallographic structures of AG7404 in complex with SARS-CoV-1 Mpro and SARS-CoV-2 Mpro and of rupintrivir in complex with SARS-CoV-2 Mpro were solved, revealing that all protein residues interacting with the inhibitors are conserved between the two proteins. A detailed analysis of protein-inhibitor interactions indicates that AG7404 has a better fit to the active site of the target protease than rupintrivir. This observation was further confirmed by biochemical FRET assays showing IC50 values of 47 µM and 101 µM for AG7404 and rupintrivir, respectively, in the case of SARS-CoV-2 Mpro. Equivalent IC50 values for SARS-CoV-1 also revealed greater inhibitory capacity of AG7404, with a value of 29 µM vs. 66 µM for rupintrivir. Finally, the antiviral activity of the two inhibitors against SARS-CoV-2 was confirmed in a human cell culture model of SARS-CoV-2 infection, although rupintrivir showed a higher potency and selectivity index in this assay.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Humanos , Antivirales/química , Cisteína Endopeptidasas/metabolismo , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Simulación del Acoplamiento Molecular
4.
Acta Crystallogr D Struct Biol ; 77(Pt 1): 11-18, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33404521

RESUMEN

Medium-resolution cryo-electron microscopy maps, in particular when they include a significant number of α-helices, may allow the building of partial models that are useful for molecular-replacement searches in large crystallographic structures when the structures of homologs are not available and experimental phasing has failed. Here, as an example, the solution of the structure of a bacteriophage portal using a partial 30% model built into a 7.8 Šresolution cryo-EM map is shown. Inspection of the self-rotation function allowed the correct oligomerization state to be determined, and density-modification procedures using rotation matrices and a mask based on the cryo-EM structure were critical for solving the structure. A workflow is described that may be applicable to similar cases and this strategy is compared with direct use of the cryo-EM map for molecular replacement.


Asunto(s)
Bacteriófago T7/metabolismo , Proteínas de la Cápside/química , Microscopía por Crioelectrón/métodos , Modelos Moleculares , Conformación Proteica , Programas Informáticos
5.
Nat Commun ; 10(1): 3891, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467275

RESUMEN

Herpesviridae is a vast family of enveloped DNA viruses that includes eight distinct human pathogens, responsible for diseases that range from almost asymptomatic to severe and life-threatening. Epstein-Barr virus infects B-cells and epithelial cells, causing infectious mononucleosis, as well as a number of cancers. Epstein-Barr infection cannot be cured since neither vaccine nor antiviral drug treatments are available. All herpesviruses contain a linear double-stranded DNA genome, enclosed within an icosahedral capsid. Viral portal protein plays a key role in the procapsid assembly and DNA packaging. The portal is the entrance and exit pore for the viral genome, making it an attractive pharmacological target for the development of new antivirals. Here we present the atomic structure of the portal protein of Epstein-Barr virus, solved by cryo-electron microscopy at 3.5 Å resolution. The detailed architecture of this protein suggests that it plays a functional role in DNA retention during packaging.


Asunto(s)
Proteínas de la Cápside/ultraestructura , Herpesvirus Humano 4/ultraestructura , Proteínas Virales/ultraestructura , Ensamble de Virus , Cápside/ultraestructura , Proteínas de la Cápside/genética , Microscopía por Crioelectrón , Empaquetamiento del ADN , ADN Viral/genética , Genoma Viral , Herpesvirus Humano 4/genética , Humanos , Modelos Moleculares , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/ultraestructura , Proteínas Virales/genética , Virión/ultraestructura
6.
Nat Commun ; 10(1): 3746, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31431626

RESUMEN

Double-stranded DNA bacteriophages package their genome at high pressure inside a procapsid through the portal, an oligomeric ring protein located at a unique capsid vertex. Once the DNA has been packaged, the tail components assemble on the portal to render the mature infective virion. The tail tightly seals the ejection conduit until infection, when its interaction with the host membrane triggers the opening of the channel and the viral genome is delivered to the host cell. Using high-resolution cryo-electron microscopy and X-ray crystallography, here we describe various structures of the T7 bacteriophage portal and fiber-less tail complex, which suggest a possible mechanism for DNA retention and ejection: a portal closed conformation temporarily retains the genome before the tail is assembled, whereas an open portal is found in the tail. Moreover, a fold including a seven-bladed ß-propeller domain is described for the nozzle tail protein.


Asunto(s)
Bacteriófago T7/fisiología , Proteínas de la Cápside/ultraestructura , Cápside/ultraestructura , Empaquetamiento del ADN , Modelos Moleculares , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , ADN Viral/metabolismo , Dominios Proteicos
7.
RNA ; 25(5): 607-619, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30737359

RESUMEN

Adenosine deaminase acting on transfer RNA (ADAT) is an essential eukaryotic enzyme that catalyzes the deamination of adenosine to inosine at the first position of tRNA anticodons. Mammalian ADATs modify eight different tRNAs, having increased their substrate range from a bacterial ancestor that likely deaminated exclusively tRNAArg Here we investigate the recognition mechanisms of tRNAArg and tRNAAla by human ADAT to shed light on the process of substrate expansion that took place during the evolution of the enzyme. We show that tRNA recognition by human ADAT does not depend on conserved identity elements, but on the overall structural features of tRNA. We find that ancestral-like interactions are conserved for tRNAArg, while eukaryote-specific substrates use alternative mechanisms. These recognition studies show that human ADAT can be inhibited by tRNA fragments in vitro, including naturally occurring fragments involved in important regulatory pathways.


Asunto(s)
Adenosina Desaminasa/metabolismo , Anticodón/química , ARN de Transferencia de Alanina/química , ARN de Transferencia de Arginina/química , Adenosina/metabolismo , Adenosina Desaminasa/genética , Anticodón/genética , Anticodón/metabolismo , Secuencia de Bases , Desaminación , Evolución Molecular , Expresión Génica , Humanos , Inosina/metabolismo , Conformación de Ácido Nucleico , ARN de Transferencia de Alanina/genética , ARN de Transferencia de Alanina/metabolismo , ARN de Transferencia de Arginina/genética , ARN de Transferencia de Arginina/metabolismo , Alineación de Secuencia , Especificidad por Sustrato
8.
ACS Omega ; 3(8): 8497-8505, 2018 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-31458978

RESUMEN

Human cytomegalovirus (HCMV) is an opportunistic pathogen causing a variety of severe viral infections, including irreversible congenital disabilities. Nowadays, HCMV infection is treated by inhibiting the viral DNA polymerase. However, DNA polymerase inhibitors have several drawbacks. An alternative strategy is to use compounds against the packaging machinery or terminase complex, which is essential for viral replication. Our discovery that raltegravir (1), a human immunodeficiency virus drug, inhibits the nuclease function of UL89, one of the protein subunits of the complex, prompted us to further develop terminase inhibitors. On the basis of the structure of 1, a library of diketoacid (α,γ-DKA and ß,δ-DKA) derivatives were synthesized and tested for UL89-C nuclease activity. The mode of action of α,γ-DKA derivatives on the UL89 active site was elucidated by using X-ray crystallography, molecular docking, and in vitro experiments. Our studies identified α,γ-DKA derivative 14 able to inhibit UL89 in vitro in the low micromolar range, making 14 an optimal candidate for further development and virus-infected cell assay.

9.
PLoS One ; 12(10): e0186286, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29045454

RESUMEN

Sulfur trafficking in living organisms relies on transpersulfuration reactions consisting in the enzyme-catalyzed transfer of S atoms via activated persulfidic S across protein-protein interfaces. The recent elucidation of the mechanistic basis for transpersulfuration in the CsdA-CsdE model system has paved the way for a better understanding of its role under oxidative stress. Herein we present the crystal structure of the oxidized, inactivated CsdE dimer at 2.4 Å resolution. The structure sheds light into the activation of the Cys61 nucleophile on its way from a solvent-secluded position in free CsdE to a fully extended conformation in the persulfurated CsdA-CsdE complex. Molecular dynamics simulations of available CsdE structures allow to delineate the sequence of conformational changes underwent by CsdE and to pinpoint the key role played by the deprotonation of the Cys61 thiol. The low-energy subunit orientation in the disulfide-bridged CsdE dimer demonstrates the likely physiologic relevance of this oxidative dead-end form of CsdE, suggesting that CsdE could act as a redox sensor in vivo.


Asunto(s)
Liasas de Carbono-Azufre/química , ARN Helicasas DEAD-box/química , Proteínas de Escherichia coli/química , Conformación Proteica , Azufre/química , Liasas de Carbono-Azufre/genética , Cristalografía por Rayos X , ARN Helicasas DEAD-box/genética , Escherichia coli/química , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Simulación de Dinámica Molecular , Estrés Oxidativo/genética , Dominios y Motivos de Interacción de Proteínas/genética , Multimerización de Proteína , Azufre/metabolismo
10.
Proc Natl Acad Sci U S A ; 114(32): E6526-E6535, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28739894

RESUMEN

Relaxases are metal-dependent nucleases that break and join DNA for the initiation and completion of conjugative bacterial gene transfer. Conjugation is the main process through which antibiotic resistance spreads among bacteria, with multidrug-resistant staphylococci and streptococci infections posing major threats to human health. The MOBV family of relaxases accounts for approximately 85% of all relaxases found in Staphylococcus aureus isolates. Here, we present six structures of the MOBV relaxase MobM from the promiscuous plasmid pMV158 in complex with several origin of transfer DNA fragments. A combined structural, biochemical, and computational approach reveals that MobM follows a previously uncharacterized histidine/metal-dependent DNA processing mechanism, which involves the formation of a covalent phosphoramidate histidine-DNA adduct for cell-to-cell transfer. We discuss how the chemical features of the high-energy phosphorus-nitrogen bond shape the dominant position of MOBV histidine relaxases among small promiscuous plasmids and their preference toward Gram-positive bacteria.


Asunto(s)
Proteínas Bacterianas/química , Roturas del ADN de Cadena Simple , ADN Bacteriano/química , Endodesoxirribonucleasas/química , Modelos Moleculares , Plásmidos/química , Staphylococcus aureus/enzimología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Histidina/química , Histidina/genética , Histidina/metabolismo , Plásmidos/genética , Plásmidos/metabolismo , Staphylococcus aureus/genética
11.
Sci Rep ; 7: 39680, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28051169

RESUMEN

Variolin B is a rare marine alkaloid that showed promising anti-cancer activity soon after its isolation. It acts as a cyclin-dependent kinase inhibitor, although the precise mechanism through which it exerts the cytotoxic effects is still unknown. The crystal structure of a variolin B bound to a DNA forming a pseudo-Holliday junction shows that this compound can also contribute, through intercalative binding, to either the formation or stabilization of multi-stranded DNA forms.


Asunto(s)
Antineoplásicos/química , Compuestos Aza/química , ADN/química , Sustancias Intercalantes/química , Pirimidinas/química , Cristalografía por Rayos X , Modelos Moleculares , Relación Estructura-Actividad
12.
Nat Commun ; 7: 12761, 2016 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-27677239

RESUMEN

Small-molecule fragments binding to biomacromolecules can be starting points for the development of drugs, but are often difficult to detect due to low affinities. Here we present a strategy that identifies protein-binding fragments through their potential to induce the target-guided formation of covalently bound, irreversible enzyme inhibitors. A protein-binding nucleophile reacts reversibly with a bis-electrophilic warhead, thereby positioning the second electrophile in close proximity of the active site of a viral protease, resulting in the covalent de-activation of the enzyme. The concept is implemented for Coxsackie virus B3 3C protease, a pharmacological target against enteroviral infections. Using an aldehyde-epoxide as bis-electrophile, active fragment combinations are validated through measuring the protein inactivation rate and by detecting covalent protein modification in mass spectrometry. The structure of one enzyme-inhibitor complex is determined by X-ray crystallography. The presented warhead activation assay provides potent non-peptidic, broad-spectrum inhibitors of enteroviral proteases.

13.
Sci Rep ; 6: 29232, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27411375

RESUMEN

The activity of RING ubiquitin ligases (E3s) depends on an interaction between the RING domain and ubiquitin conjugating enzymes (E2), but posttranslational events or additional structural elements, yet largely undefined, are frequently required to enhance or regulate activity. Here, we show for the ubiquitin ligase RNF125 that, in addition to the RING domain, a C2HC Zn finger (ZnF) is crucial for activity, and a short linker sequence (Li2(120-128)) enhances activity. The contribution of these regions was first shown with truncated proteins, and the essential role of the ZnF was confirmed with mutations at the Zn chelating Cys residues. Using NMR, we established that the C2HC ZnF/Li2(120-128) region is crucial for binding of the RING domain to the E2 UbcH5a. The partial X-ray structure of RNF125 revealed the presence of extensive intramolecular interactions between the RING and C2HC ZnF. A mutation at one of the contact residues in the C2HC ZnF, a highly conserved M112, resulted in the loss of ubiquitin ligase activity. Thus, we identified the structural basis for an essential role of the C2HC ZnF and conclude that this domain stabilizes the RING domain, and is therefore required for binding of RNF125 to an E2.


Asunto(s)
Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Células HEK293 , Humanos , Modelos Moleculares , Proteínas Mutantes/metabolismo , Mutación Puntual/genética , Unión Proteica , Dominios Proteicos , Relación Estructura-Actividad , Ubiquitina/metabolismo , Dedos de Zinc
14.
Sci Rep ; 6: 20915, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26875695

RESUMEN

DNA replication initiation is a vital and tightly regulated step in all replicons and requires an initiator factor that specifically recognizes the DNA replication origin and starts replication. RepB from the promiscuous streptococcal plasmid pMV158 is a hexameric ring protein evolutionary related to viral initiators. Here we explore the conformational plasticity of the RepB hexamer by i) SAXS, ii) sedimentation experiments, iii) molecular simulations and iv) X-ray crystallography. Combining these techniques, we derive an estimate of the conformational ensemble in solution showing that the C-terminal oligomerisation domains of the protein form a rigid cylindrical scaffold to which the N-terminal DNA-binding/catalytic domains are attached as highly flexible appendages, featuring multiple orientations. In addition, we show that the hinge region connecting both domains plays a pivotal role in the observed plasticity. Sequence comparisons and a literature survey show that this hinge region could exists in other initiators, suggesting that it is a common, crucial structural element for DNA binding and manipulation.


Asunto(s)
ADN Helicasas/química , Replicación del ADN/genética , Proteínas de Unión al ADN/química , Conformación de Ácido Nucleico , Secuencia de Aminoácidos/genética , Cristalografía por Rayos X , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Simulación de Dinámica Molecular , Plásmidos/genética , Dominios Proteicos , Multimerización de Proteína , Origen de Réplica/genética , Streptococcus/genética
16.
PLoS One ; 10(4): e0118606, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25897750

RESUMEN

Cyclic N6-threonylcarbamoyladenosine ('cyclic t6A', ct(6)A) is a non-thiolated hypermodification found in transfer RNAs (tRNAs) in bacteria, protists, fungi and plants. In bacteria and yeast cells ct(6)A has been shown to enhance translation fidelity and efficiency of ANN codons by improving the faithful discrimination of aminoacylated tRNAs by the ribosome. To further the understanding of ct(6)A biology we have determined the high-resolution crystal structures of CsdL/TcdA in complex with AMP and ATP, an E1-like activating enzyme from Escherichia coli, which catalyzes the ATP-dependent dehydration of t6A to form ct(6)A. CsdL/TcdA is a dimer whose structural integrity and dimer interface depend critically on strongly bound K+ and Na+ cations. By using biochemical assays and small-angle X-ray scattering we show that CsdL/TcdA can associate with tRNA with a 1:1 stoichiometry and with the proper position and orientation for the cyclization of t6A. Furthermore, we show by nuclear magnetic resonance that CsdL/TcdA engages in transient interactions with CsdA and CsdE, which, in the latter case, involve catalytically important residues. These short-lived interactions may underpin the precise channeling of sulfur atoms from cysteine to CsdL/TcdA as previously characterized. In summary, the combination of structural, biophysical and biochemical methods applied to CsdL/TcdA has afforded a more thorough understanding of how the structure of this E1-like enzyme has been fine tuned to accomplish ct(6)A synthesis on tRNAs while providing support for the notion that CsdA and CsdE are able to functionally interact with CsdL/TcdA.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Nucleotidiltransferasas/química , Nucleotidiltransferasas/metabolismo , ARN de Transferencia/metabolismo , Enzimas Activadoras de Ubiquitina/química , Enzimas Activadoras de Ubiquitina/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Escherichia coli/química , Modelos Moleculares , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Conformación Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
17.
J Cell Sci ; 128(9): 1824-34, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25908846

RESUMEN

Tubulin proteostasis is regulated by a group of molecular chaperones termed tubulin cofactors (TBC). Whereas tubulin heterodimer formation is well-characterized biochemically, its dissociation pathway is not clearly understood. Here, we carried out biochemical assays to dissect the role of the human TBCE and TBCB chaperones in α-tubulin-ß-tubulin dissociation. We used electron microscopy and image processing to determine the three-dimensional structure of the human TBCE, TBCB and α-tubulin (αEB) complex, which is formed upon α-tubulin-ß-tubulin heterodimer dissociation by the two chaperones. Docking the atomic structures of domains of these proteins, including the TBCE UBL domain, as we determined by X-ray crystallography, allowed description of the molecular architecture of the αEB complex. We found that heterodimer dissociation is an energy-independent process that takes place through a disruption of the α-tubulin-ß-tubulin interface that is caused by a steric interaction between ß-tubulin and the TBCE cytoskeleton-associated protein glycine-rich (CAP-Gly) and leucine-rich repeat (LRR) domains. The protruding arrangement of chaperone ubiquitin-like (UBL) domains in the αEB complex suggests that there is a direct interaction of this complex with the proteasome, thus mediating α-tubulin degradation.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Chaperonas Moleculares/metabolismo , Multimerización de Proteína , Tubulina (Proteína)/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Bovinos , Cristalografía por Rayos X , Proteínas Fluorescentes Verdes/metabolismo , Guanosina Trifosfato , Humanos , Hidrólisis , Proteínas Asociadas a Microtúbulos/química , Modelos Biológicos , Modelos Moleculares , Chaperonas Moleculares/química , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteolisis , Tubulina (Proteína)/química
18.
PLoS One ; 9(7): e102139, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25014014

RESUMEN

In order to maintain proper cellular function, the metabolism of the bacterial microbiota presents several mechanisms oriented to keep a correctly balanced amino acid pool. Central components of these mechanisms are enzymes with alanine transaminase activity, pyridoxal 5'-phosphate-dependent enzymes that interconvert alanine and pyruvate, thereby allowing the precise control of alanine and glutamate concentrations, two of the most abundant amino acids in the cellular amino acid pool. Here we report the 2.11-Å crystal structure of full-length AlaA from the model organism Escherichia coli, a major bacterial alanine aminotransferase, and compare its overall structure and active site composition with detailed atomic models of two other bacterial enzymes capable of catalyzing this reaction in vivo, AlaC and valine-pyruvate aminotransferase (AvtA). Apart from a narrow entry channel to the active site, a feature of this new crystal structure is the role of an active site loop that closes in upon binding of substrate-mimicking molecules, and which has only been previously reported in a plant enzyme. Comparison of the available structures indicates that beyond superficial differences, alanine aminotransferases of diverse phylogenetic origins share a universal reaction mechanism that depends on an array of highly conserved amino acid residues and is similarly regulated by various unrelated motifs. Despite this unifying mechanism and regulation, growth competition experiments demonstrate that AlaA, AlaC and AvtA are not freely exchangeable in vivo, suggesting that their functional repertoire is not completely redundant thus providing an explanation for their independent evolutionary conservation.


Asunto(s)
Alanina Transaminasa/química , Alanina/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Ácido Pirúvico/química , Transaminasas/química , Alanina/metabolismo , Alanina Transaminasa/genética , Alanina Transaminasa/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Secuencia Conservada , Cristalografía por Rayos X , Escherichia coli/química , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Evolución Molecular , Expresión Génica , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Estructura Secundaria de Proteína , Ácido Pirúvico/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología Estructural de Proteína , Especificidad por Sustrato , Transaminasas/genética , Transaminasas/metabolismo
19.
Cell ; 156(3): 577-89, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24485461

RESUMEN

Auxin regulates numerous plant developmental processes by controlling gene expression via a family of functionally distinct DNA-binding auxin response factors (ARFs), yet the mechanistic basis for generating specificity in auxin response is unknown. Here, we address this question by solving high-resolution crystal structures of the pivotal Arabidopsis developmental regulator ARF5/MONOPTEROS (MP), its divergent paralog ARF1, and a complex of ARF1 and a generic auxin response DNA element (AuxRE). We show that ARF DNA-binding domains also homodimerize to generate cooperative DNA binding, which is critical for in vivo ARF5/MP function. Strikingly, DNA-contacting residues are conserved between ARFs, and we discover that monomers have the same intrinsic specificity. ARF1 and ARF5 homodimers, however, differ in spacing tolerated between binding sites. Our data identify the DNA-binding domain as an ARF dimerization domain, suggest that ARF dimers bind complex sites as molecular calipers with ARF-specific spacing preference, and provide an atomic-scale mechanistic model for specificity in auxin response.


Asunto(s)
Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Ácidos Indolacéticos/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , ADN/química , Dimerización , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Estructura Terciaria de Proteína , Alineación de Secuencia
20.
Angew Chem Int Ed Engl ; 53(7): 1949-52, 2014 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-24449275

RESUMEN

The crystal structure of the Δ,Δ enantiomer of the binuclear "light-switch" ruthenium complex [µ-(11,11'-bidppz)(1,10-phenanthroline)4 Ru2 ](4+) bound to the oligonucleotide d(CGTACG) shows that one dppz moiety of the dumbbell-like compound inserts into the DNA stack through the extrusion of an AT base pair. The second dppz moiety recruits a neighboring DNA molecule, and the complex thus cross-links two adjacent duplexes by bridging their major grooves.


Asunto(s)
Complejos de Coordinación/química , ADN/química , Sustancias Intercalantes/química , Rutenio/química , Emparejamiento Base , Modelos Moleculares , Fenazinas/química , Piridonas/química , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...