Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 52(12): 7188-7210, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38686810

RESUMO

Genome-wide approaches have significantly advanced our knowledge of the repertoire of RNA-binding proteins (RBPs) that associate with cellular polyadenylated mRNAs within eukaryotic cells. Recent studies focusing on the RBP interactomes of viral mRNAs, notably SARS-Cov-2, have revealed both similarities and differences between the RBP profiles of viral and cellular mRNAs. However, the RBPome of influenza virus mRNAs remains unexplored. Herein, we identify RBPs that associate with the viral mRNA encoding the nucleoprotein (NP) of an influenza A virus. Focusing on TDP-43, we show that it binds several influenza mRNAs beyond the NP-mRNA, and that its depletion results in lower levels of viral mRNAs and proteins within infected cells, and a decreased yield of infectious viral particles. We provide evidence that the viral polymerase recruits TDP-43 onto viral mRNAs through a direct interaction with the disordered C-terminal domain of TDP-43. Notably, other RBPs found to be associated with influenza virus mRNAs also interact with the viral polymerase, which points to a role of the polymerase in orchestrating the assembly of viral messenger ribonucleoproteins.


Assuntos
Proteínas de Ligação a DNA , Vírus da Influenza A , RNA Mensageiro , RNA Viral , Proteínas de Ligação a RNA , Replicação Viral , Humanos , Replicação Viral/genética , RNA Viral/metabolismo , RNA Viral/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Vírus da Influenza A/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Proteínas do Nucleocapsídeo/genética , Células HEK293 , Proteínas do Core Viral/metabolismo , Proteínas do Core Viral/genética , Ligação Proteica , Animais
2.
J Am Chem Soc ; 145(2): 800-810, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36599057

RESUMO

Prodrugs have little or no pharmacological activity and are converted to active drugs in the body by enzymes, metabolic reactions, or through human-controlled actions. However, prodrugs promoting their chemical bioconversion without any of these processes have not been reported before. Here, we present an enzyme-independent prodrug activation mechanism by boron-based compounds (benzoxaboroles) targeting leucyl-tRNA synthetase (LeuRS), including an antibiotic that recently has completed phase II clinical trials to cure tuberculosis. We combine nuclear magnetic resonance spectroscopy and X-ray crystallography with isothermal titration calorimetry to show that these benzoxaboroles do not bind directly to their drug target LeuRS, instead they are prodrugs that activate their bioconversion by forming a highly specific and reversible LeuRS inhibition adduct with ATP, AMP, or the terminal adenosine of the tRNALeu. We demonstrate how the oxaborole group of the prodrugs cyclizes with the adenosine ribose at physiological concentrations to form the active molecule. This bioconversion mechanism explains the remarkably good druglike properties of benzoxaboroles showing efficacy against radically different human pathogens and fully explains the mechanism of action of these compounds. Thus, this adenosine-dependent activation mechanism represents a novel concept in prodrug chemistry that can be applied to improve the solubility, permeability and metabolic stability of challenging drugs.


Assuntos
Aminoacil-tRNA Sintetases , Leucina-tRNA Ligase , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Adenosina/farmacologia , Leucina-tRNA Ligase/genética , Antibacterianos/farmacologia
3.
Nucleic Acids Res ; 48(9): 4946-4959, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32232361

RESUMO

Human cytosolic leucyl-tRNA synthetase (hcLRS) is an essential and multifunctional enzyme. Its canonical function is to catalyze the covalent ligation of leucine to tRNALeu, and it may also hydrolyze mischarged tRNAs through an editing mechanism. Together with eight other aminoacyl-tRNA synthetases (AaRSs) and three auxiliary proteins, it forms a large multi-synthetase complex (MSC). Beyond its role in translation, hcLRS has an important moonlight function as a leucine sensor in the rapamycin complex 1 (mTORC1) pathway. Since this pathway is active in cancer development, hcLRS is a potential target for anti-tumor drug development. Moreover, LRS from pathogenic microbes are proven drug targets for developing antibiotics, which however should not inhibit hcLRS. Here we present the crystal structure of hcLRS at a 2.5 Å resolution, the first complete structure of a eukaryotic LRS, and analyze the binding of various compounds that target different sites of hcLRS. We also deduce the assembly mechanism of hcLRS into the MSC through reconstitution of the entire mega complex in vitro. Overall, our study provides the molecular basis for understanding both the multifaceted functions of hcLRS and for drug development targeting these functions.


Assuntos
Leucina-tRNA Ligase/química , Anti-Infecciosos/química , Biocatálise , Domínio Catalítico , Desenho de Fármacos , Humanos , Leucina-tRNA Ligase/efeitos dos fármacos , Leucina-tRNA Ligase/metabolismo , Modelos Moleculares , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Domínios Proteicos , RNA de Transferência de Leucina/metabolismo , Aminoacilação de RNA de Transferência
4.
Nat Commun ; 9(1): 1820, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29739942

RESUMO

RIG-I is a viral RNA sensor that induces the production of type I interferon (IFN) in response to infection with a variety of viruses. Modification of RIG-I with K63-linked poly-ubiquitin chains, synthesised by TRIM25, is crucial for activation of the RIG-I/MAVS signalling pathway. TRIM25 activity is targeted by influenza A virus non-structural protein 1 (NS1) to suppress IFN production and prevent an efficient host immune response. Here we present structures of the human TRIM25 coiled-coil-PRYSPRY module and of complexes between the TRIM25 coiled-coil domain and NS1. These structures show that binding of NS1 interferes with the correct positioning of the PRYSPRY domain of TRIM25 required for substrate ubiquitination and provide a mechanistic explanation for how NS1 suppresses RIG-I ubiquitination and hence downstream signalling. In contrast, the formation of unanchored K63-linked poly-ubiquitin chains is unchanged by NS1 binding, indicating that RING dimerisation of TRIM25 is not affected by NS1.


Assuntos
Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/imunologia , Proteínas com Motivo Tripartido/antagonistas & inibidores , Proteínas com Motivo Tripartido/imunologia , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/imunologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/imunologia , Sequência de Aminoácidos , Células Cultivadas , Proteína DEAD-box 58/imunologia , Células HEK293 , Humanos , Interferons/biossíntese , Ligação Proteica , Domínios Proteicos , Multimerização Proteica , RNA Viral/imunologia , Receptores Imunológicos , Transdução de Sinais , Fatores de Transcrição/química , Proteínas com Motivo Tripartido/química , Ubiquitina-Proteína Ligases/química , Ubiquitinação
5.
Nat Commun ; 7: 12928, 2016 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-27713402

RESUMO

Antibiotic-producing microbes evolved self-resistance mechanisms to avoid suicide. The biocontrol Agrobacterium radiobacter K84 secretes the Trojan Horse antibiotic agrocin 84 that is selectively transported into the plant pathogen A. tumefaciens and processed into the toxin TM84. We previously showed that TM84 employs a unique tRNA-dependent mechanism to inhibit leucyl-tRNA synthetase (LeuRS), while the TM84-producer prevents self-poisoning by expressing a resistant LeuRS AgnB2. We now identify a mechanism by which the antibiotic-producing microbe resists its own toxin. Using a combination of structural, biochemical and biophysical approaches, we show that AgnB2 evolved structural changes so as to resist the antibiotic by eliminating the tRNA-dependence of TM84 binding. Mutagenesis of key resistance determinants results in mutants adopting an antibiotic-sensitive phenotype. This study illuminates the evolution of resistance in self-immunity genes and provides mechanistic insights into a fascinating tRNA-dependent antibiotic with applications for the development of anti-infectives and the prevention of biocontrol emasculation.


Assuntos
Agrobacterium tumefaciens/genética , Farmacorresistência Bacteriana/genética , Evolução Molecular , Leucina-tRNA Ligase/antagonistas & inibidores , Leucina-tRNA Ligase/genética , Genes Bacterianos , Cinética , Leucina-tRNA Ligase/metabolismo , Controle Biológico de Vetores , Tumores de Planta/microbiologia , RNA de Transferência/metabolismo
6.
Mol Cell ; 62(4): 586-602, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27203181

RESUMO

RIG-I and MDA5 sense virus-derived short 5'ppp blunt-ended or long dsRNA, respectively, causing interferon production. Non-signaling LGP2 appears to positively and negatively regulate MDA5 and RIG-I signaling, respectively. Co-crystal structures of chicken (ch) LGP2 with dsRNA display a fully or semi-closed conformation depending on the presence or absence of nucleotide. LGP2 caps blunt, 3' or 5' overhang dsRNA ends with 1 bp longer overall footprint than RIG-I. Structures of 1:1 and 2:1 complexes of chMDA5 with short dsRNA reveal head-to-head packing rather than the polar head-to-tail orientation described for long filaments. chLGP2 and chMDA5 make filaments with a similar axial repeat, although less co-operatively for chLGP2. Overall, LGP2 resembles a chimera combining a MDA5-like helicase domain and RIG-I like CTD supporting both stem and end binding. Functionally, RNA binding is required for LGP2-mediated enhancement of MDA5 activation. We propose that LGP2 end-binding may promote nucleation of MDA5 oligomerization on dsRNA.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas Aviárias/metabolismo , Proteína DEAD-box 58/metabolismo , Helicase IFIH1 Induzida por Interferon/metabolismo , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Difosfato de Adenosina/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Trifosfato de Adenosina/metabolismo , Animais , Proteínas Aviárias/química , Proteínas Aviárias/genética , Sítios de Ligação , Linhagem Celular , Galinhas , Proteína DEAD-box 58/química , Proteína DEAD-box 58/genética , Humanos , Hidrólise , Helicase IFIH1 Induzida por Interferon/química , Helicase IFIH1 Induzida por Interferon/genética , Modelos Moleculares , Conformação de Ácido Nucleico , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/genética , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Receptores de Reconhecimento de Padrão/química , Receptores de Reconhecimento de Padrão/genética , Relação Estrutura-Atividade , Transfecção
7.
Sci Rep ; 6: 24727, 2016 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-27095520

RESUMO

The genome of influenza A virus (IAV) comprises eight RNA segments (vRNA) which are transcribed and replicated by the heterotrimeric IAV RNA-dependent RNA-polymerase (RdRp). RdRp consists of three subunits (PA, PB1 and PB2) and binds both the highly conserved 3'- and 5'-ends of the vRNA segment. The IAV RdRp is an important antiviral target, but its structural mechanism has remained largely elusive to date. By applying a polyprotein strategy, we produced RdRp complexes and define a minimal human IAV RdRp core complex. We show that PA-PB1 forms a stable heterodimeric submodule that can strongly interact with 5'-vRNA. In contrast, 3'-vRNA recognition critically depends on the PB2 N-terminal domain. Moreover, we demonstrate that PA-PB1 forms a stable and stoichiometric complex with host nuclear import factor RanBP5 that can be modelled using SAXS and we show that the PA-PB1-RanPB5 complex is no longer capable of 5'-vRNA binding. Our results provide further evidence for a step-wise assembly of IAV structural components, regulated by nuclear transport mechanisms and host factor binding.


Assuntos
Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Influenza Humana/metabolismo , Influenza Humana/virologia , Subunidades Proteicas/metabolismo , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , beta Carioferinas/metabolismo , Regulação Viral da Expressão Gênica , Humanos , Regiões Promotoras Genéticas , Ligação Proteica , Multimerização Proteica , RNA Viral/genética , RNA Polimerase Dependente de RNA/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
8.
J Phys Chem B ; 120(19): 4388-98, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27115861

RESUMO

Leucyl-tRNA synthetase catalyzes attachment of leucine amino acid to its cognate tRNA. During the second, aminoacetylation, step of the reaction, the leucyl moiety is transferred from leucyl-adenylate to the terminal A76 adenosine of tRNA. In this work, we have investigated the aminoacetylation step catalyzed by leucyl-tRNA synthase, using ab initio quantum chemical/molecular mechanical hybrid potentials in conjunction with reaction-path-location algorithms and molecular dynamics free energy simulations. We have modeled reaction mechanisms arising from both crystallographic studies and computational work. We invoke various groups as potential proton acceptors-namely, the phosphate and leucyl amino groups of leucyl-adenylate, the A76 base of tRNA, and the Asp80 and Glu532 residues of the protein-and consider both metal-assisted and metal-free reactions. Free energy calculations indicate that both the phosphate group of leucyl adenylate and Glu532 are not strong bases. This agrees with the results of the quantum chemical/molecular mechanical reaction path calculations which give high free energy barriers for the studied pathways involving these groups. A self-assisted mechanism with the leucyl amino group and Asp80 as proton acceptors is the most likely. Furthermore, in this mechanism the presence of a metal ion coordinated by the phosphate group and Glu532 strongly activates the reaction.


Assuntos
Leucina-tRNA Ligase/metabolismo , Acetilação , Biocatálise , Cristalografia por Raios X , Escherichia coli/enzimologia , Leucina-tRNA Ligase/química , Metais/química , Simulação de Dinâmica Molecular , Estrutura Terciária de Proteína , Teoria Quântica , Especificidade por Substrato , Termodinâmica , Água/química , Água/metabolismo
9.
Nature ; 527(7576): 114-7, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26503046

RESUMO

Negative-sense RNA viruses, such as influenza, encode large, multidomain RNA-dependent RNA polymerases that can both transcribe and replicate the viral RNA genome. In influenza virus, the polymerase (FluPol) is composed of three polypeptides: PB1, PB2 and PA/P3. PB1 houses the polymerase active site, whereas PB2 and PA/P3 contain, respectively, cap-binding and endonuclease domains required for transcription initiation by cap-snatching. Replication occurs through de novo initiation and involves a complementary RNA intermediate. Currently available structures of the influenza A and B virus polymerases include promoter RNA (the 5' and 3' termini of viral genome segments), showing FluPol in transcription pre-initiation states. Here we report the structure of apo-FluPol from an influenza C virus, solved by X-ray crystallography to 3.9 Å, revealing a new 'closed' conformation. The apo-FluPol forms a compact particle with PB1 at its centre, capped on one face by PB2 and clamped between the two globular domains of P3. Notably, this structure is radically different from those of promoter-bound FluPols. The endonuclease domain of P3 and the domains within the carboxy-terminal two-thirds of PB2 are completely rearranged. The cap-binding site is occluded by PB2, resulting in a conformation that is incompatible with transcription initiation. Thus, our structure captures FluPol in a closed, transcription pre-activation state. This reveals the conformation of newly made apo-FluPol in an infected cell, but may also apply to FluPol in the context of a non-transcribing ribonucleoprotein complex. Comparison of the apo-FluPol structure with those of promoter-bound FluPols allows us to propose a mechanism for FluPol activation. Our study demonstrates the remarkable flexibility of influenza virus RNA polymerase, and aids our understanding of the mechanisms controlling transcription and genome replication.


Assuntos
Gammainfluenzavirus/enzimologia , RNA Polimerase Dependente de RNA/química , Apoenzimas/química , Apoenzimas/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Endonucleases/química , Endonucleases/metabolismo , Ativação Enzimática , Modelos Moleculares , Iniciação Traducional da Cadeia Peptídica , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Capuzes de RNA/metabolismo , RNA Viral/biossíntese , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Ribonucleoproteínas/química
10.
BMC Biol ; 13: 54, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26215161

RESUMO

BACKGROUND: The cytoplasmic RIG-like receptors are responsible for the early detection of viruses and other intracellular microbes by activating the innate immune response mediated by type I interferons (IFNs). RIG-I and MDA5 detect virus-specific RNA motifs with short 5'-tri/diphosphorylated, blunt-end double-stranded RNA (dsRNA) and >0.5-2 kb long dsRNA as canonical agonists, respectively. However, in vitro, they can bind to many RNA species, while in cells there is an activation threshold. As SF2 helicase/ATPase family members, ATP hydrolysis is dependent on co-operative RNA and ATP binding. Whereas simultaneous ATP and cognate RNA binding is sufficient to activate RIG-I by releasing autoinhibition of the signaling domains, the physiological role of the ATPase activity of RIG-I and MDA5 remains controversial. RESULTS: A cross-analysis of a rationally designed panel of RNA binding and ATPase mutants and truncated receptors, using type I IFN promoter activation as readout, allows us to refine our understanding of the structure-function relationships of RIG-I and MDA5. RNA activation of RIG-I depends on multiple critical RNA binding sites in its helicase domain as confirmed by functional evidence using novel mutations. We found that RIG-I or MDA5 mutants with low ATP hydrolysis activity exhibit constitutive activity but this was fully reverted when associated with mutations preventing RNA binding to the helicase domain. We propose that the turnover kinetics of the ATPase domain enables the discrimination of self/non-self RNA by both RIG-I and MDA5. Non-cognate, possibly self, RNA binding would lead to fast ATP turnover and RNA disassociation and thus insufficient time for the caspase activation and recruitment domains (CARDs) to promote downstream signaling, whereas tighter cognate RNA binding provides a longer time window for downstream events to be engaged. CONCLUSIONS: The exquisite fine-tuning of RIG-I and MDA5 RNA-dependent ATPase activity coupled to CARD release allows a robust IFN response from a minor subset of non-self RNAs within a sea of cellular self RNAs. This avoids the eventuality of deleterious autoimmunity effects as have been recently described to arise from natural gain-of-function alleles of RIG-I and MDA5.


Assuntos
Adenosina Trifosfatases/metabolismo , RNA Helicases DEAD-box/metabolismo , RNA de Cadeia Dupla/metabolismo , Adenosina Trifosfatases/genética , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Humanos , Hidrólise , Helicase IFIH1 Induzida por Interferon , Queratina-8/metabolismo , Cinética , Mutação Puntual , Ligação Proteica , Estrutura Terciária de Proteína , RNA de Cadeia Dupla/química , Receptores Imunológicos
11.
PLoS One ; 9(9): e108770, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25259935

RESUMO

Effective host defence against viruses depends on the rapid triggering of innate immunity through the induction of a type I interferon (IFN) response. To this end, microbe-associated molecular patterns are detected by dedicated receptors. Among them, the RIG-I-like receptors RIG-I and MDA5 activate IFN gene expression upon sensing viral RNA in the cytoplasm. While MDA5 forms long filaments in vitro upon activation, RIG-I is believed to oligomerize after RNA binding in order to transduce a signal. Here, we show that in vitro binding of synthetic RNA mimicking that of Mononegavirales (Ebola, rabies and measles viruses) leader sequences to purified RIG-I does not induce RIG-I oligomerization. Furthermore, in cells devoid of endogenous functional RIG-I-like receptors, after activation of exogenous Flag-RIG-I by a 62-mer-5'ppp-dsRNA or by polyinosinic:polycytidylic acid, a dsRNA analogue, or by measles virus infection, anti-Flag immunoprecipitation and specific elution with Flag peptide indicated a monomeric form of RIG-I. Accordingly, when using the Gaussia Luciferase-Based Protein Complementation Assay (PCA), a more sensitive in cellula assay, no RIG-I oligomerization could be detected upon RNA stimulation. Altogether our data indicate that the need for self-oligomerization of RIG-I for signal transduction is either dispensable or very transient.


Assuntos
RNA Helicases DEAD-box/metabolismo , Imunidade Inata/fisiologia , Interferon beta/genética , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteína DEAD-box 58 , Células HEK293 , Humanos , Regiões Promotoras Genéticas , RNA Viral/genética , Receptores Imunológicos , Células Vero
12.
Cell ; 157(7): 1698-711, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24910301

RESUMO

Germline-specific Piwi-interacting RNAs (piRNAs) protect animal genomes against transposons and are essential for fertility. piRNAs targeting active transposons are amplified by the ping-pong cycle, which couples Piwi endonucleolytic slicing of target RNAs to biogenesis of new piRNAs. Here, we describe the identification of a transient Amplifier complex that mediates biogenesis of secondary piRNAs in insect cells. Amplifier is nucleated by the DEAD box RNA helicase Vasa and contains the two Piwi proteins participating in the ping-pong loop, the Tudor protein Qin/Kumo and antisense piRNA guides. These components assemble on the surface of Vasa's helicase domain, which functions as an RNA clamp to anchor Amplifier onto transposon transcripts. We show that ATP-dependent RNP remodeling by Vasa facilitates transfer of 5' sliced piRNA precursors between ping-pong partners, and loss of this activity causes sterility in Drosophila. Our results reveal the molecular basis for the small RNA amplification that confers adaptive immunity against transposons.


Assuntos
Bombyx/metabolismo , Proteínas de Insetos/metabolismo , RNA Interferente Pequeno/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Bombyx/genética , Linhagem Celular , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Elementos de DNA Transponíveis , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Proteínas de Insetos/genética , Mutação , Ovário/citologia , Ovário/metabolismo
13.
Structure ; 22(3): 452-65, 2014 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-24607143

RESUMO

Vaccinia virus capping enzyme is a heterodimer of D1 (844 aa) and D12 (287 aa) polypeptides that executes all three steps in m(7)GpppRNA synthesis. The D1 subunit comprises an N-terminal RNA triphosphatase (TPase)-guanylyltransferase (GTase) module and a C-terminal guanine-N7-methyltransferase (MTase) module. The D12 subunit binds and allosterically stimulates the MTase module. Crystal structures of the complete D1⋅D12 heterodimer disclose the TPase and GTase as members of the triphosphate tunnel metalloenzyme and covalent nucleotidyltransferase superfamilies, respectively, albeit with distinctive active site features. An extensive TPase-GTase interface clamps the GTase nucleotidyltransferase and OB-fold domains in a closed conformation around GTP. Mutagenesis confirms the importance of the TPase-GTase interface for GTase activity. The D1⋅D12 structure complements and rationalizes four decades of biochemical studies of this enzyme, which was the first capping enzyme to be purified and characterized, and provides new insights into the origins of the capping systems of other large DNA viruses.


Assuntos
Metiltransferases/química , Metiltransferases/metabolismo , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Monoéster Fosfórico Hidrolases/química , Monoéster Fosfórico Hidrolases/metabolismo , Vaccinia virus/enzimologia , Domínio Catalítico , Cristalografia por Raios X , Evolução Molecular , Metiltransferases/genética , Modelos Moleculares , Complexos Multienzimáticos/genética , Mutação , Nucleotidiltransferases/genética , Monoéster Fosfórico Hidrolases/genética , Conformação Proteica , Multimerização Proteica , Proteínas Virais
14.
Cell Rep ; 5(1): 13-20, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24095733

RESUMO

PRDM9, a histone lysine methyltransferase, is a key determinant of the localization of meiotic recombination hot spots in humans and mice and the only vertebrate protein known to be involved in hybrid sterility. Here, we report the crystal structure of the PRDM9 methyltransferase domain in complex with a histone H3 peptide dimethylated on lysine 4 (H3K4me2) and S-adenosylhomocysteine (AdoHcy), which provides insights into the methyltransferase activity of PRDM proteins. We show that the genuine substrate of PRDM9 is histone H3 lysine 4 (H3K4) and that the enzyme possesses mono-, di-, and trimethylation activities. We also determined the crystal structure of PRDM9 in its autoinhibited state, which revealed a rearrangement of the substrate and cofactor binding sites by a concerted action of the pre-SET and post-SET domains, providing important insights into the regulatory mechanisms of histone lysine methyltransferase activity.


Assuntos
Histona-Lisina N-Metiltransferase/química , Sequência de Aminoácidos , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Alinhamento de Sequência , Especificidade por Substrato
15.
Proc Natl Acad Sci U S A ; 110(10): 3817-22, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23431144

RESUMO

Mycoplasma leucyl-tRNA synthetases (LeuRSs) have been identified in which the connective polypeptide 1 (CP1) amino acid editing domain that clears mischarged tRNAs are missing (Mycoplasma mobile) or highly degenerate (Mycoplasma synoviae). Thus, these enzymes rely on a clearance pathway called pretransfer editing, which hydrolyzes misactivated aminoacyl-adenylate intermediate via a nebulous mechanism that has been controversial for decades. Even as the sole fidelity pathway for clearing amino acid selection errors in the pathogenic M. mobile, pretransfer editing is not robust enough to completely block mischarging of tRNA(Leu), resulting in codon ambiguity and statistical proteins. A high-resolution X-ray crystal structure shows that M. mobile LeuRS structurally overlaps with other LeuRS cores. However, when CP1 domains from different aminoacyl-tRNA synthetases and origins were fused to this common LeuRS core, surprisingly, pretransfer editing was enhanced. It is hypothesized that the CP1 domain evolved as a molecular rheostat to balance multiple functions. These include distal control of specificity and enzyme activity in the ancient canonical core, as well as providing a separate hydrolytic active site for clearing mischarged tRNA.


Assuntos
Leucina-tRNA Ligase/química , Leucina-tRNA Ligase/metabolismo , Mycoplasma/genética , Mycoplasma/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Códon/genética , Códon/metabolismo , Cristalografia por Raios X , Leucina-tRNA Ligase/genética , Modelos Moleculares , Dados de Sequência Molecular , Mycoplasma/patogenicidade , Mycoplasma synoviae/enzimologia , Mycoplasma synoviae/genética , Conformação Proteica , Estrutura Terciária de Proteína , Edição de RNA , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , RNA de Transferência de Leucina/genética , RNA de Transferência de Leucina/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos
16.
Nat Commun ; 4: 1417, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23361008

RESUMO

Leucyl-tRNA synthetases (LeuRSs) have an essential role in translation and are promising targets for antibiotic development. Agrocin 84 is a LeuRS inhibitor produced by the biocontrol agent Agrobacterium radiobacter K84 that targets pathogenic strains of A. tumefaciens, the causative agent of plant tumours. Agrocin 84 acts as a molecular Trojan horse and is processed inside the pathogen into a toxic moiety (TM84). Here we show using crystal structure, thermodynamic and kinetic analyses, that this natural antibiotic employs a unique and previously undescribed mechanism to inhibit LeuRS. TM84 requires tRNA(Leu) for tight binding to the LeuRS synthetic active site, unlike any previously reported inhibitors. TM84 traps the enzyme-tRNA complex in a novel 'aminoacylation-like' conformation, forming novel interactions with the KMSKS loop and the tRNA 3'-end. Our findings reveal an intriguing tRNA-dependent inhibition mechanism that may confer a distinct evolutionary advantage in vivo and inform future rational antibiotic design.


Assuntos
Nucleotídeos de Adenina/farmacologia , Agrobacterium tumefaciens/enzimologia , Agentes de Controle Biológico , Leucina-tRNA Ligase/antagonistas & inibidores , Tumores de Planta/microbiologia , RNA de Plantas/metabolismo , RNA de Transferência/metabolismo , Nucleotídeos de Adenina/química , Agrobacterium tumefaciens/efeitos dos fármacos , Aminoacilação/efeitos dos fármacos , Calorimetria , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Leucina-tRNA Ligase/química , Leucina-tRNA Ligase/metabolismo , Modelos Moleculares , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Estrutura Terciária de Proteína , RNA de Plantas/química , RNA de Transferência/química
17.
RNA ; 18(12): 2118-27, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23118418

RESUMO

A series of high-resolution crystal structures of RIG-I and RIG-I:dsRNA cocrystals has recently been reported. Comparison of these structures provides considerable insight into how this innate immune pattern recognition receptor is activated upon detecting and binding a certain class of viral RNAs.


Assuntos
RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/metabolismo , Cristalografia por Raios X , Proteína DEAD-box 58 , RNA Helicases DEAD-box/imunologia , Ativação Enzimática , Humanos , Imunidade Inata , Ligantes , Substâncias Macromoleculares , Modelos Biológicos , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/metabolismo , RNA Viral/química , RNA Viral/metabolismo , Receptores Imunológicos , Transdução de Sinais
18.
PLoS One ; 7(9): e46075, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23049939

RESUMO

During the viral life cycle adenoviruses produce excess capsid proteins. Human adenovirus serotype 3 (Ad3) synthesizes predominantly an excess of free pentons, the complexes of pentameric penton base and trimeric fiber proteins, which are responsible for virus penetration. In infected cells Ad3 pentons spontaneously assemble into dodecahedral virus-like nano-particles containing twelve pentons. They also form in insect cells during expression in the baculovirus system. Similarly, in the absence of fiber protein dodecahedric particles built of 12 penton base pentamers can be produced. Both kinds of dodecahedra show remarkable efficiency of intracellular penetration and can be engineered to deliver several millions of foreign cargo molecules to a single target cell. For this reason, they are of great interest as a delivery vector. In order to successfully manipulate this potential vector for drug and/or gene delivery, an understanding of the molecular basis of vector assembly and integrity is critical. Crystallographic data in conjunction with site-directed mutagenesis and biochemical analysis provide a model for the molecular determinants of dodecamer particle assembly and the requirements for stability. The 3.8 Å crystal structure of Ad3 penton base dodecamer (Dd) shows that the dodecahedric structure is stabilized by strand-swapping between neighboring penton base molecules. Such N-terminal strand-swapping does not occur for Dd of Ad2, a serotype which does not form Dd under physiological conditions. This unique stabilization of the Ad3 dodecamer is controlled by residues 59-61 located at the site of strand switching, the residues involved in putative salt bridges between pentamers and by the disordered N-terminus (residues 1-47), as confirmed by site directed mutagenesis and biochemical analysis of mutant and wild type protein. We also provide evidence that the distal N-terminal residues are externally exposed and available for attaching cargo.


Assuntos
Adenovírus Humanos/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Sequência de Aminoácidos , Proteínas do Capsídeo/genética , Cristalografia por Raios X , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Secundária de Proteína , Homologia de Sequência de Aminoácidos
19.
Cell ; 147(2): 423-35, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-22000019

RESUMO

RIG-I is a key innate immune pattern-recognition receptor that triggers interferon expression upon detection of intracellular 5'triphosphate double-stranded RNA (5'ppp-dsRNA) of viral origin. RIG-I comprises N-terminal caspase activation and recruitment domains (CARDs), a DECH helicase, and a C-terminal domain (CTD). We present crystal structures of the ligand-free, autorepressed, and RNA-bound, activated states of RIG-I. Inactive RIG-I has an open conformation with the CARDs sequestered by a helical domain inserted between the two helicase moieties. ATP and dsRNA binding induce a major rearrangement to a closed conformation in which the helicase and CTD bind the blunt end 5'ppp-dsRNA with perfect complementarity but incompatibly with continued CARD binding. We propose that after initial binding of 5'ppp-dsRNA to the flexibly linked CTD, co-operative tight binding of ATP and RNA to the helicase domain liberates the CARDs for downstream signaling. These findings significantly advance our molecular understanding of the activation of innate immune signaling helicases.


Assuntos
Patos/metabolismo , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo , Receptores de Reconhecimento de Padrão/química , Receptores do Ácido Retinoico/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Galinhas/imunologia , Patos/imunologia , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína , RNA de Cadeia Dupla/imunologia , RNA Viral/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Receptores do Ácido Retinoico/química , Receptores do Ácido Retinoico/imunologia
20.
J Med Chem ; 52(12): 3666-78, 2009 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-19456100

RESUMO

The adenovirus serotype Ad37 binds to and infects human corneal epithelial (HCE) cells through attachment to cellular glycoproteins carrying terminal sialic acids. By use of the crystallographic structure of the sialic acid-interacting domain of the Ad37 fiber protein in complex with sialyllactose, a set of N-acyl modified sialic acids were designed to improve binding affinity through increased hydrophobic interactions. These N-acyl modified sialic acids and their corresponding multivalent human serum albumin (HSA) conjugates were synthesized and tested in Ad37 cell binding and cell infectivity assays. Compounds bearing small substituents were as effective inhibitors as sialic acid. X-ray crystallography and overlays with the Ad37-sialyllactose complex showed that the N-acyl modified sialic acids were positioned in the same orientation as sialic acid. Their multivalent counterparts achieved a strong multivalency effect and were more effective to prevent infection than the monomers. Unfortunately, they were less active as inhibitors than multivalent sialic acid.


Assuntos
Adenovírus Humanos/efeitos dos fármacos , Desenho de Fármacos , Ceratoconjuntivite/epidemiologia , Ceratoconjuntivite/virologia , Ácidos Siálicos/síntese química , Ácidos Siálicos/farmacologia , Sítios de Ligação , Simulação por Computador , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Humanos , Modelos Químicos , Modelos Moleculares , Conformação Molecular , Albumina Sérica/química , Ácidos Siálicos/química , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA