Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(11): 1336-1345, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32887977

RESUMEN

The development of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines and therapeutics will depend on understanding viral immunity. We studied T cell memory in 42 patients following recovery from COVID-19 (28 with mild disease and 14 with severe disease) and 16 unexposed donors, using interferon-γ-based assays with peptides spanning SARS-CoV-2 except ORF1. The breadth and magnitude of T cell responses were significantly higher in severe as compared with mild cases. Total and spike-specific T cell responses correlated with spike-specific antibody responses. We identified 41 peptides containing CD4+ and/or CD8+ epitopes, including six immunodominant regions. Six optimized CD8+ epitopes were defined, with peptide-MHC pentamer-positive cells displaying the central and effector memory phenotype. In mild cases, higher proportions of SARS-CoV-2-specific CD8+ T cells were observed. The identification of T cell responses associated with milder disease will support an understanding of protective immunity and highlights the potential of including non-spike proteins within future COVID-19 vaccine design.


Asunto(s)
Antígenos Virales/inmunología , Betacoronavirus/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , COVID-19 , Vacunas contra la COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/prevención & control , Epítopos de Linfocito T/inmunología , Humanos , Epítopos Inmunodominantes/inmunología , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/patología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/inmunología , Reino Unido , Vacunas Virales/inmunología
2.
Nature ; 617(7960): 409-416, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37138077

RESUMEN

CrAssphage and related viruses of the order Crassvirales (hereafter referred to as crassviruses) were originally discovered by cross-assembly of metagenomic sequences. They are the most abundant viruses in the human gut, are found in the majority of individual gut viromes, and account for up to 95% of the viral sequences in some individuals1-4. Crassviruses are likely to have major roles in shaping the composition and functionality of the human microbiome, but the structures and roles of most of the virally encoded proteins are unknown, with only generic predictions resulting from bioinformatic analyses4,5. Here we present a cryo-electron microscopy reconstruction of Bacteroides intestinalis virus ΦcrAss0016, providing the structural basis for the functional assignment of most of its virion proteins. The muzzle protein forms an assembly about 1 MDa in size at the end of the tail and exhibits a previously unknown fold that we designate the 'crass fold', that is likely to serve as a gatekeeper that controls the ejection of cargos. In addition to packing the approximately 103 kb of virus DNA, the ΦcrAss001 virion has extensive storage space for virally encoded cargo proteins in the capsid and, unusually, within the tail. One of the cargo proteins is present in both the capsid and the tail, suggesting a general mechanism for protein ejection, which involves partial unfolding of proteins during their extrusion through the tail. These findings provide a structural basis for understanding the mechanisms of assembly and infection of these highly abundant crassviruses.


Asunto(s)
Virus ADN , Intestinos , Proteínas Virales , Virión , Humanos , Cápside/química , Cápside/metabolismo , Cápside/ultraestructura , Microscopía por Crioelectrón , Virus ADN/química , Virus ADN/clasificación , Virus ADN/aislamiento & purificación , Virus ADN/metabolismo , Virus ADN/ultraestructura , Virión/química , Virión/metabolismo , Virión/ultraestructura , Ensamble de Virus , Intestinos/microbiología , Intestinos/virología , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Virales/ultraestructura , Desplegamiento Proteico , Pliegue de Proteína
3.
Nucleic Acids Res ; 51(13): 7025-7035, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37293963

RESUMEN

Double-stranded DNA viruses utilise machinery, made of terminase proteins, to package viral DNA into the capsid. For cos bacteriophage, a defined signal, recognised by small terminase, flanks each genome unit. Here we present the first structural data for a cos virus DNA packaging motor, assembled from the bacteriophage HK97 terminase proteins, procapsids encompassing the portal protein, and DNA containing a cos site. The cryo-EM structure is consistent with the packaging termination state adopted after DNA cleavage, with DNA density within the large terminase assembly ending abruptly at the portal protein entrance. Retention of the large terminase complex after cleavage of the short DNA substrate suggests that motor dissociation from the capsid requires headful pressure, in common with pac viruses. Interestingly, the clip domain of the 12-subunit portal protein does not adhere to C12 symmetry, indicating asymmetry induced by binding of the large terminase/DNA. The motor assembly is also highly asymmetric, showing a ring of 5 large terminase monomers, tilted against the portal. Variable degrees of extension between N- and C-terminal domains of individual subunits suggest a mechanism of DNA translocation driven by inter-domain contraction and relaxation.


Asunto(s)
Bacteriófagos , Ensamble de Virus , Bacteriófagos/genética , Bacteriófagos/metabolismo , Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/química , Empaquetamiento del ADN , ADN Viral/genética , Endodesoxirribonucleasas/metabolismo
4.
Subcell Biochem ; 104: 181-205, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38963488

RESUMEN

Tailed double-stranded DNA bacteriophage employs a protein terminase motor to package their genome into a preformed protein shell-a system shared with eukaryotic dsDNA viruses such as herpesviruses. DNA packaging motor proteins represent excellent targets for antiviral therapy, with Letermovir, which binds Cytomegalovirus terminase, already licensed as an effective prophylaxis. In the realm of bacterial viruses, these DNA packaging motors comprise three protein constituents: the portal protein, small terminase and large terminase. The portal protein guards the passage of DNA into the preformed protein shell and acts as a protein interaction hub throughout viral assembly. Small terminase recognises the viral DNA and recruits large terminase, which in turn pumps DNA in an ATP-dependent manner. Large terminase also cleaves DNA at the termination of packaging. Multiple high-resolution structures of each component have been resolved for different phages, but it is only more recently that the field has moved towards cryo-EM reconstructions of protein complexes. In conjunction with highly informative single-particle studies of packaging kinetics, these structures have begun to inspire models for the packaging process and its place among other DNA machines.


Asunto(s)
ADN Viral , Proteínas Virales , ADN Viral/genética , ADN Viral/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/genética , Endodesoxirribonucleasas/metabolismo , Endodesoxirribonucleasas/genética , Empaquetamiento del Genoma Viral/fisiología , Empaquetamiento del ADN , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/metabolismo , Genoma Viral
5.
Nucleic Acids Res ; 50(15): 8719-8732, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-35947691

RESUMEN

Many essential cellular processes rely on substrate rotation or translocation by a multi-subunit, ring-type NTPase. A large number of double-stranded DNA viruses, including tailed bacteriophages and herpes viruses, use a homomeric ring ATPase to processively translocate viral genomic DNA into procapsids during assembly. Our current understanding of viral DNA packaging comes from three archetypal bacteriophage systems: cos, pac and phi29. Detailed mechanistic understanding exists for pac and phi29, but not for cos. Here, we reconstituted in vitro a cos packaging system based on bacteriophage HK97 and provided a detailed biochemical and structural description. We used a photobleaching-based, single-molecule assay to determine the stoichiometry of the DNA-translocating ATPase large terminase. Crystal structures of the large terminase and DNA-recruiting small terminase, a first for a biochemically defined cos system, reveal mechanistic similarities between cos and pac systems. At the same time, mutational and biochemical analyses indicate a new regulatory mechanism for ATPase multimerization and coordination in the HK97 system. This work therefore establishes a framework for studying the evolutionary relationships between ATP-dependent DNA translocation machineries in double-stranded DNA viruses.


Asunto(s)
Adenosina Trifosfatasas , Ensamble de Virus , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/química , Ensamble de Virus/genética , Proteínas Virales/genética , Proteínas Virales/química , Empaquetamiento del ADN , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/química , ADN Viral/genética , ADN Viral/química
6.
PLoS Pathog ; 17(7): e1009740, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34270629

RESUMEN

Nipah and its close relative Hendra are highly pathogenic zoonotic viruses, storing their ssRNA genome in a helical nucleocapsid assembly formed by the N protein, a major viral immunogen. Here, we report the first cryoEM structure for a Henipavirus RNA-bound nucleocapsid assembly, at 3.5 Å resolution. The helical assembly is stabilised by previously undefined N- and C-terminal segments, contributing to subunit-subunit interactions. RNA is wrapped around the nucleocapsid protein assembly with a periodicity of six nucleotides per protomer, in the "3-bases-in, 3-bases-out" conformation, with protein plasticity enabling non-sequence specific interactions. The structure reveals commonalities in RNA binding pockets and in the conformation of bound RNA, not only with members of the Paramyxoviridae family, but also with the evolutionarily distant Filoviridae Ebola virus. Significant structural differences with other Paramyxoviridae members are also observed, particularly in the position and length of the exposed α-helix, residues 123-139, which may serve as a valuable epitope for surveillance and diagnostics.


Asunto(s)
Virus Nipah/ultraestructura , Proteínas de la Nucleocápside/ultraestructura , Nucleocápside/ultraestructura , Microscopía por Crioelectrón , Modelos Moleculares , Conformación Molecular , Virus Nipah/química , Nucleocápside/química , Proteínas de la Nucleocápside/química , ARN Viral/química , ARN Viral/ultraestructura
7.
J Am Chem Soc ; 144(49): 22540-22548, 2022 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-36455212

RESUMEN

The application of nanopores as label-free, single-molecule biosensors for electrical or optical probing of structural features in biomolecules has been widely explored. While biological nanopores (membrane proteins and bacteriophage portal proteins) and solid-state nanopores (thin films and two-dimensional materials) have been extensively employed, the third class of nanopores known as hybrid nanopores, where an artificial membrane substitutes the organic support membrane of proteins, has been only sparsely studied due to challenges in implementation. G20c portal protein contains a natural DNA pore that is used by viruses for filling their capsid with viral genomic DNA. We have previously developed a lipid-free hybrid nanopore by "corking" the G20c portal protein into a SiNx nanopore. Herein, we demonstrate that through chemical functionalization of the synthetic nanopore, covalent linkage between the solid-state pore and the G20c portal protein considerably improves the hybrid pore stability, lifetime, and voltage resilience. Moreover, we demonstrate electric-field-driven and motor protein-mediated transport of DNA molecules through this hybrid nanopore. Our integrated protein/solid-state device can serve as a robust and durable framework for sensing and sequencing at high voltages, potentially providing higher resolution, higher signal-to-noise ratio, and higher throughput compared to the more conventional membrane-embedded protein platforms.


Asunto(s)
Bacteriófagos , Técnicas Biosensibles , Nanoporos , Nanotecnología/métodos , ADN Viral
8.
Proc Natl Acad Sci U S A ; 116(9): 3556-3561, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30737287

RESUMEN

Double-stranded DNA viruses, including bacteriophages and herpesviruses, package their genomes into preformed capsids, using ATP-driven motors. Seeking to advance structural and mechanistic understanding, we established in vitro packaging for a thermostable bacteriophage, P23-45 of Thermus thermophilus Both the unexpanded procapsid and the expanded mature capsid can package DNA in the presence of packaging ATPase over the 20 °C to 70 °C temperature range, with optimum activity at 50 °C to 65 °C. Cryo-EM reconstructions for the mature and immature capsids at 3.7-Å and 4.4-Å resolution, respectively, reveal conformational changes during capsid expansion. Capsomer interactions in the expanded capsid are reinforced by formation of intersubunit ß-sheets with N-terminal segments of auxiliary protein trimers. Unexpectedly, the capsid has T=7 quasi-symmetry, despite the P23-45 genome being twice as large as those of known T=7 phages, in which the DNA is compacted to near-crystalline density. Our data explain this anomaly, showing how the canonical HK97 fold has adapted to double the volume of the capsid, while maintaining its structural integrity. Reconstructions of the procapsid and the expanded capsid defined the structure of the single vertex containing the portal protein. Together with a 1.95-Å resolution crystal structure of the portal protein and DNA packaging assays, these reconstructions indicate that capsid expansion affects the conformation of the portal protein, while still allowing DNA to be packaged. These observations suggest a mechanism by which structural events inside the capsid can be communicated to the outside.


Asunto(s)
Bacteriófagos/ultraestructura , Cápside/ultraestructura , Empaquetamiento del ADN/genética , Virus ADN/ultraestructura , Bacteriófagos/genética , Microscopía por Crioelectrón , Virus ADN/genética , ADN Viral/genética , ADN Viral/ultraestructura , Virión/genética , Virión/ultraestructura , Ensamble de Virus/genética
9.
Nucleic Acids Res ; 47(6): 3208-3222, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30698796

RESUMEN

Pif1 is a multifunctional helicase and DNA processing enzyme that has roles in genome stability. The enzyme is conserved in eukaryotes and also found in some prokaryotes. The functions of human PIF1 (hPIF1) are also critical for survival of certain tumour cell lines during replication stress, making it an important target for cancer therapy. Crystal structures of hPIF1 presented here explore structural events along the chemical reaction coordinate of ATP hydrolysis at an unprecedented level of detail. The structures for the apo as well as the ground and transition states reveal conformational adjustments in defined protein segments that can trigger larger domain movements required for helicase action. Comparisons with the structures of yeast and bacterial Pif1 reveal a conserved ssDNA binding channel in hPIF1 that we show is critical for single-stranded DNA binding during unwinding, but not the binding of G quadruplex DNA. Mutational analysis suggests that while the ssDNA-binding channel is important for helicase activity, it is not used in DNA annealing. Structural differences, in particular in the DNA strand separation wedge region, highlight significant evolutionary divergence of the human PIF1 protein from bacterial and yeast orthologues.


Asunto(s)
ADN Helicasas/química , ADN de Cadena Simple/química , Proteínas de Unión al ADN/química , Nucleótidos/química , Adenosina Trifosfato/química , Adenosina Trifosfato/genética , Cristalografía por Rayos X , ADN Helicasas/genética , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/genética , Inestabilidad Genómica , Humanos , Hidrólisis , Nucleótidos/genética , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/química
10.
Nucleic Acids Res ; 45(22): 13029-13042, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29069443

RESUMEN

The crystal structure of the large terminase from the Geobacillus stearothermophilus bacteriophage D6E shows a unique relative orientation of the N-terminal adenosine triphosphatase (ATPase) and C-terminal nuclease domains. This monomeric 'initiation' state with the two domains 'locked' together is stabilized via a conserved C-terminal arm, which may interact with the portal protein during motor assembly, as predicted for several bacteriophages. Further work supports the formation of an active oligomeric state: (i) AUC data demonstrate the presence of oligomers; (ii) mutational analysis reveals a trans-arginine finger, R158, indispensable for ATP hydrolysis; (iii) the location of this arginine is conserved with the HerA/FtsK ATPase superfamily; (iv) a molecular docking model of the pentamer is compatible with the location of the identified arginine finger. However, this pentameric model is structurally incompatible with the monomeric 'initiation' state and is supported by the observed increase in kcat of ATP hydrolysis, from 7.8 ± 0.1 min-1 to 457.7 ± 9.2 min-1 upon removal of the C-terminal nuclease domain. Taken together, these structural, biophysical and biochemical data suggest a model where transition from the 'initiation' state into a catalytically competent pentameric state, is accompanied by substantial domain rearrangements, triggered by the removal of the C-terminal arm from the ATPase active site.


Asunto(s)
Adenosina Trifosfato/metabolismo , Bacteriófagos/enzimología , Endodesoxirribonucleasas/metabolismo , Proteínas Virales/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Bacteriófagos/genética , Cristalografía por Rayos X , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/genética , Geobacillus stearothermophilus/virología , Calor , Hidrólisis , Modelos Moleculares , Mutación , Conformación Proteica , Multimerización de Proteína , Homología de Secuencia de Aminoácido , Proteínas Virales/química , Proteínas Virales/genética
11.
Nucleic Acids Res ; 45(6): 3580-3590, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28100693

RESUMEN

Bacteriophages and large dsDNA viruses encode sophisticated machinery to translocate their DNA into a preformed empty capsid. An essential part of this machine, the large terminase protein, processes viral DNA into constituent units utilizing its nuclease activity. Crystal structures of the large terminase nuclease from the thermophilic bacteriophage G20c show that it is most similar to the RuvC family of the RNase H-like endonucleases. Like RuvC proteins, the nuclease requires either Mn2+, Mg2+ or Co2+ ions for activity, but is inactive with Zn2+ and Ca2+. High resolution crystal structures of complexes with different metals reveal that in the absence of DNA, only one catalytic metal ion is accommodated in the active site. Binding of the second metal ion may be facilitated by conformational variability, which enables the two catalytic aspartic acids to be brought closer to each other. Structural comparison indicates that in common with the RuvC family, the location of the two catalytic metals differs from other members of the RNase H family. In contrast to a recently proposed mechanism, the available data do not support binding of the two metals at an ultra-short interatomic distance. Thus we postulate that viral terminases cleave DNA by the canonical RuvC-like mechanism.


Asunto(s)
Endodesoxirribonucleasas/química , Metales/química , Proteínas Virales/química , Biocatálisis , Dominio Catalítico , División del ADN , ADN Viral/metabolismo , Endodesoxirribonucleasas/metabolismo , Genoma Viral , Modelos Moleculares , Recombinasas/química , Thermus thermophilus/enzimología , Proteínas Virales/metabolismo , Ensamble de Virus
12.
Nucleic Acids Res ; 44(2): 776-89, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26673721

RESUMEN

The helix-turn-helix (HTH) motif features frequently in protein DNA-binding assemblies. Viral pac site-targeting small terminase proteins possess an unusual architecture in which the HTH motifs are displayed in a ring, distinct from the classical HTH dimer. Here we investigate how such a circular array of HTH motifs enables specific recognition of the viral genome for initiation of DNA packaging during virus assembly. We found, by surface plasmon resonance and analytical ultracentrifugation, that individual HTH motifs of the Bacillus phage SF6 small terminase bind the packaging regions of SF6 and related SPP1 genome weakly, with little local sequence specificity. Nuclear magnetic resonance chemical shift perturbation studies with an arbitrary single-site substrate suggest that the HTH motif contacts DNA similarly to how certain HTH proteins contact DNA non-specifically. Our observations support a model where specificity is generated through conformational selection of an intrinsically bent DNA segment by a ring of HTHs which bind weakly but cooperatively. Such a system would enable viral gene regulation and control of the viral life cycle, with a minimal genome, conferring a major evolutionary advantage for SPP1-like viruses.


Asunto(s)
Fagos de Bacillus/genética , Endodesoxirribonucleasas/metabolismo , Ensamble de Virus/fisiología , Fagos de Bacillus/fisiología , Sitios de Unión , ADN/química , ADN/metabolismo , Empaquetamiento del ADN , ADN Viral/química , ADN Viral/metabolismo , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/genética , Secuencias Hélice-Giro-Hélice , Modelos Moleculares , Mutación , Conformación Proteica , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie , Ultracentrifugación/métodos , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo , Ensamble de Virus/genética
13.
Proc Natl Acad Sci U S A ; 112(19): 6033-7, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25902496

RESUMEN

The reduction of specific uridines to dihydrouridine is one of the most common modifications in tRNA. Increased levels of the dihydrouridine modification are associated with cancer. Dihydrouridine synthases (Dus) from different subfamilies selectively reduce distinct uridines, located at spatially unique positions of folded tRNA, into dihydrouridine. Because the catalytic center of all Dus enzymes is conserved, it is unclear how the same protein fold can be reprogrammed to ensure that nucleotides exposed at spatially distinct faces of tRNA can be accommodated in the same active site. We show that the Escherichia coli DusC is specific toward U16 of tRNA. Unexpectedly, crystal structures of DusC complexes with tRNA(Phe) and tRNA(Trp) show that Dus subfamilies that selectively modify U16 or U20 in tRNA adopt identical folds but bind their respective tRNA substrates in an almost reverse orientation that differs by a 160° rotation. The tRNA docking orientation appears to be guided by subfamily-specific clusters of amino acids ("binding signatures") together with differences in the shape of the positively charged tRNA-binding surfaces. tRNA orientations are further constrained by positional differences between the C-terminal "recognition" domains. The exquisite substrate specificity of Dus enzymes is therefore controlled by a relatively simple mechanism involving major reorientation of the whole tRNA molecule. Such reprogramming of the enzymatic specificity appears to be a unique evolutionary solution for altering tRNA recognition by the same protein fold.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Oxidorreductasas/química , ARN de Transferencia/química , Aminoácidos/química , Dominio Catalítico , Cristalografía por Rayos X , Evolución Molecular , Unión Proteica , Pliegue de Proteína , ARN/química , Proteínas de Unión al ARN/química , Especificidad por Sustrato , Uridina/química , Difracción de Rayos X
14.
Nucleic Acids Res ; 43(12): 6009-22, 2015 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-26013813

RESUMEN

Base Excision Repair (BER) efficiently corrects the most common types of DNA damage in mammalian cells. Step-by-step coordination of BER is facilitated by multiple interactions between enzymes and accessory proteins involved. Here we characterize quantitatively a number of complexes formed by DNA polymerase ß (Polß), apurinic/apyrimidinic endonuclease 1 (APE1), poly(ADP-ribose) polymerase 1 (PARP1), X-ray repair cross-complementing protein 1 (XRCC1) and tyrosyl-DNA phosphodiesterase 1 (TDP1), using fluorescence- and light scattering-based techniques. Direct physical interactions between the APE1-Polß, APE1-TDP1, APE1-PARP1 and Polß-TDP1 pairs have been detected and characterized for the first time. The combined results provide strong evidence that the most stable complex is formed between XRCC1 and Polß. Model DNA intermediates of BER are shown to induce significant rearrangement of the Polß complexes with XRCC1 and PARP1, while having no detectable influence on the protein-protein binding affinities. The strength of APE1 interaction with Polß, XRCC1 and PARP1 is revealed to be modulated by BER intermediates to different extents, depending on the type of DNA damage. The affinity of APE1 for Polß is higher in the complex with abasic site-containing DNA than after the APE1-catalyzed incision. Our findings advance understanding of the molecular mechanisms underlying coordination and regulation of the BER process.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Animales , ADN/metabolismo , ADN Polimerasa beta/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Fluorescencia , Transferencia Resonante de Energía de Fluorescencia , Humanos , Luz , Hidrolasas Diéster Fosfóricas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ratas , Dispersión de Radiación , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
15.
Dev Dyn ; 245(1): 34-46, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26447465

RESUMEN

BACKGROUND: Lin28 proteins are post-transcriptional regulators of gene expression with multiple roles in development and the regulation of pluripotency in stem cells. Much attention has focussed on Lin28 proteins as negative regulators of let-7 miRNA biogenesis; a function that is conserved in several animal groups and in multiple processes. However, there is increasing evidence that Lin28 proteins have additional roles, distinct from regulation of let-7 abundance. We have previously demonstrated that lin28 proteins have functions associated with the regulation of early cell lineage specification in Xenopus embryos, independent of a lin28/let-7 regulatory axis. However, the nature of lin28 targets in Xenopus development remains obscure. RESULTS: Here, we show that mir-17∼92 and mir-106∼363 cluster miRNAs are down-regulated in response to lin28 knockdown, and RNAs from these clusters are co-expressed with lin28 genes during germ layer specification. Mature miRNAs derived from pre-mir-363 are most sensitive to lin28 inhibition. We demonstrate that lin28a binds to the terminal loop of pre-mir-363 with an affinity similar to that of let-7, and that this high affinity interaction requires to conserved a GGAG motif. CONCLUSIONS: Our data suggest a novel function for amphibian lin28 proteins as positive regulators of mir-17∼92 family miRNAs.


Asunto(s)
Desarrollo Embrionario/genética , Regulación de la Expresión Génica , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Embrión no Mamífero/fisiología , Estratos Germinativos/metabolismo , MicroARNs/genética , Proteínas de Unión al ARN/genética , Xenopus , Proteínas de Xenopus/genética
16.
Biochemistry ; 55(36): 5021-7, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27559824

RESUMEN

Lin28A is a post-transcriptional regulator of gene expression that interacts with and negatively regulates the biogenesis of let-7 family miRNAs. Recent data suggested that Lin28A also binds the putative tumor suppressor miR-363, a member of the 106~363 cluster of miRNAs. Affinity for this miRNA and the stoichiometry of the protein-RNA complex are unknown. Characterization of human Lin28's interaction with RNA has been complicated by difficulties in producing stable RNA-free protein. We have engineered a maltose binding protein fusion with Lin28, which binds let-7 miRNA with a Kd of 54.1 ± 4.2 nM, in agreement with previous data on a murine homologue. We show that human Lin28A binds miR-363 with a 1:1 stoichiometry and with a similar, if not higher, affinity (Kd = 16.6 ± 1.9 nM). Further analysis suggests that the interaction of the N-terminal cold shock domain of Lin28A with RNA is salt-dependent, supporting a model in which the cold shock domain allows the protein to sample RNA substrates through transient electrostatic interactions.


Asunto(s)
MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Polarización de Fluorescencia , Humanos , Unión Proteica
17.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 7): 1564-71, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26143927

RESUMEN

The reduction of uridine to dihydrouridine at specific positions in tRNA is catalysed by dihydrouridine synthase (Dus) enzymes. Increased expression of human dihydrouridine synthase 2 (hDus2) has been linked to pulmonary carcinogenesis, while its knockdown decreased cancer cell line viability, suggesting that it may serve as a valuable target for therapeutic intervention. Here, the X-ray crystal structure of a construct of hDus2 encompassing the catalytic and tRNA-recognition domains (residues 1-340) determined at 1.9 Šresolution is presented. It is shown that the structure can be determined automatically by phenix.mr_rosetta starting from a bacterial Dus enzyme with only 18% sequence identity and a significantly divergent structure. The overall fold of the human Dus2 is similar to that of bacterial enzymes, but has a larger recognition domain and a unique three-stranded antiparallel ß-sheet insertion into the catalytic domain that packs next to the recognition domain, contributing to domain-domain interactions. The structure may inform the development of novel therapeutic approaches in the fight against lung cancer.


Asunto(s)
Oxidorreductasas/química , Bacterias/química , Bacterias/enzimología , Bacterias/metabolismo , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Neoplasias Pulmonares/enzimología , Modelos Moleculares , Oxidorreductasas/metabolismo , Conformación Proteica , ARN de Transferencia/metabolismo
18.
Proc Natl Acad Sci U S A ; 109(3): 811-6, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22207627

RESUMEN

Genome packaging into preformed viral procapsids is driven by powerful molecular motors. The small terminase protein is essential for the initial recognition of viral DNA and regulates the motor's ATPase and nuclease activities during DNA translocation. The crystal structure of a full-length small terminase protein from the Siphoviridae bacteriophage SF6, comprising the N-terminal DNA binding, the oligomerization core, and the C-terminal ß-barrel domains, reveals a nine-subunit circular assembly in which the DNA-binding domains are arranged around the oligomerization core in a highly flexible manner. Mass spectrometry analysis and four further crystal structures show that, although the full-length protein exclusively forms nine-subunit assemblies, protein constructs missing the C-terminal ß-barrel form both nine-subunit and ten-subunit assemblies, indicating the importance of the C terminus for defining the oligomeric state. The mechanism by which a ring-shaped small terminase oligomer binds viral DNA has not previously been elucidated. Here, we probed binding in vitro by using EPR and surface plasmon resonance experiments, which indicated that interaction with DNA is mediated exclusively by the DNA-binding domains and suggested a nucleosome-like model in which DNA binds around the outside of the protein oligomer.


Asunto(s)
ADN/metabolismo , Proteínas Motoras Moleculares/química , Siphoviridae/fisiología , Ensamble de Virus/fisiología , ADN/química , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/metabolismo , Espectrometría de Masas , Modelos Moleculares , Proteínas Motoras Moleculares/metabolismo , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Eliminación de Secuencia , Siphoviridae/enzimología
19.
Nucleic Acids Res ; 40(5): 2271-83, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22067453

RESUMEN

The mechanism of DNA translocation by papillomavirus E1 and polyomavirus LTag hexameric helicases involves consecutive remodelling of subunit-subunit interactions around the hexameric ring. Our biochemical analysis of E1 helicase demonstrates that a 26-residue C-terminal segment is critical for maintaining the hexameric assembly. As this segment was not resolved in previous crystallographic analysis of E1 and LTag hexameric helicases, we determined the solution structure of the intact hexameric E1 helicase by Small Angle X-ray Scattering. We find that the C-terminal segment is flexible and occupies a cleft between adjacent subunits in the ring. Electrostatic potential calculations indicate that the negatively charged C-terminus can bridge the positive electrostatic potentials of adjacent subunits. Our observations support a model in which the C-terminal peptide serves as a flexible 'brace' maintaining the oligomeric state during conformational changes associated with ATP hydrolysis. We argue that these interactions impart processivity to DNA unwinding. Sequence and disorder analysis suggest that this mechanism of hexamer stabilization would be conserved among papillomavirus E1 and polyomavirus LTag hexameric helicases.


Asunto(s)
ADN Helicasas/química , Proteínas de Unión al ADN/química , Proteínas Virales/química , Secuencia de Aminoácidos , Secuencia Conservada , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Dispersión del Ángulo Pequeño , Eliminación de Secuencia , Electricidad Estática , Proteínas Virales/genética , Proteínas Virales/metabolismo , Difracción de Rayos X
20.
bioRxiv ; 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38915634

RESUMEN

Single-stranded DNA bacteriophages of the Microviridae family are major components of the global virosphere. Microviruses are highly abundant in aquatic ecosystems and are prominent members of the mammalian gut microbiome, where their diversity has been linked to various chronic health disorders. Despite the clear importance of microviruses, little is known about the molecular mechanism of host infection. Here, we have characterized an exceptionally large microvirus, Ebor, and provide crucial insights into long-standing mechanistic questions. Cryogenic electron microscopy of Ebor revealed a capsid with trimeric protrusions that recognise lipopolysaccharides on the host surface. Cryogenic electron tomography of the host cell colonized with virus particles demonstrated that the virus initially attaches to the cell via five such protrusions, located at the corners of a single pentamer. This interaction triggers a stargate mechanism of capsid opening along the 5-fold symmetry axis, enabling delivery of the virus genome. Despite variations in specific virus-host interactions among different Microviridae family viruses, structural data indicate that the stargate mechanism of infection is universally employed by all members of the family. Startlingly, our data reveal a mechanistic link for the opening of relatively small capsids made out of a single jelly-roll fold with the structurally unrelated giant viruses.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA