Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Nature ; 607(7917): 191-196, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35732732

RESUMEN

Bacterial conjugation is the fundamental process of unidirectional transfer of DNAs, often plasmid DNAs, from a donor cell to a recipient cell1. It is the primary means by which antibiotic resistance genes spread among bacterial populations2,3. In Gram-negative bacteria, conjugation is mediated by a large transport apparatus-the conjugative type IV secretion system (T4SS)-produced by the donor cell and embedded in both its outer and inner membranes. The T4SS also elaborates a long extracellular filament-the conjugative pilus-that is essential for DNA transfer4,5. Here we present a high-resolution cryo-electron microscopy (cryo-EM) structure of a 2.8 megadalton T4SS complex composed of 92 polypeptides representing 8 of the 10 essential T4SS components involved in pilus biogenesis. We added the two remaining components to the structural model using co-evolution analysis of protein interfaces, to enable the reconstitution of the entire system including the pilus. This structure describes the exceptionally large protein-protein interaction network required to assemble the many components that constitute a T4SS and provides insights on the unique mechanism by which they elaborate pili.


Asunto(s)
Proteínas Bacterianas , Microscopía por Crioelectrón , Sistemas de Secreción Tipo IV , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Conjugación Genética , ADN/genética , Evolución Molecular , Fimbrias Bacterianas/metabolismo , Plásmidos/genética , Sistemas de Secreción Tipo IV/química , Sistemas de Secreción Tipo IV/metabolismo , Sistemas de Secreción Tipo IV/ultraestructura
2.
J Biol Chem ; 298(12): 102662, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36334629

RESUMEN

Fibrillization of the protein amyloid ß is assumed to trigger Alzheimer's pathology. Approaches that target amyloid plaques, however, have garnered limited clinical success, and their failures may relate to the scarce understanding of the impact of potential drugs on the intertwined stages of fibrillization. Here, we demonstrate that bexarotene, a T-cell lymphoma medication with known antiamyloid activity both in vitro and in vivo, suppresses amyloid fibrillization by promoting an alternative fibril structure. We employ time-resolved in situ atomic force microscopy to quantify the kinetics of growth of individual fibrils and supplement it with structure characterization by cryo-EM. We show that fibrils with structure engineered by the drug nucleate and grow substantially slower than "normal" fibrils; remarkably, growth remains stunted even in drug-free solutions. We find that the suppression of fibril growth by bexarotene is not because of the drug binding to the fibril tips or to the peptides in the solution. Kinetic analyses attribute the slow growth of drug-enforced fibril polymorph to the distinctive dynamics of peptide chain association to their tips. As an additional benefit, the bexarotene fibrils kill primary rat hippocampal neurons less efficiently than normal fibrils. In conclusion, the suggested drug-driven polymorph transformation presents a mode of action to irreversibly suppress toxic aggregates not only in Alzheimer's but also potentially in myriad diverse pathologies that originate with protein condensation.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Animales , Ratas , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/metabolismo , Bexaroteno/farmacología , Amiloide/química , Placa Amiloide , Fragmentos de Péptidos/química
3.
EMBO J ; 36(20): 3080-3095, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-28923826

RESUMEN

Type IV secretion (T4S) systems are versatile bacterial secretion systems mediating transport of protein and/or DNA T4S systems are generally composed of 11 VirB proteins and 1 VirD protein (VirD4). The VirB1-11 proteins assemble to form a secretion machinery and a pilus while the VirD4 protein is responsible for substrate recruitment. The structure of VirD4 in isolation is known; however, its structure bound to the VirB1-11 apparatus has not been determined. Here, we purify a T4S system with VirD4 bound, define the biochemical requirements for complex formation and describe the protein-protein interaction network in which VirD4 is involved. We also solve the structure of this complex by negative stain electron microscopy, demonstrating that two copies of VirD4 dimers locate on both sides of the apparatus, in between the VirB4 ATPases. Given the central role of VirD4 in type IV secretion, our study provides mechanistic insights on a process that mediates the dangerous spread of antibiotic resistance genes among bacterial populations.


Asunto(s)
Agrobacterium tumefaciens/ultraestructura , Sustancias Macromoleculares/aislamiento & purificación , Sustancias Macromoleculares/ultraestructura , Sistemas de Secreción Tipo IV/aislamiento & purificación , Sistemas de Secreción Tipo IV/ultraestructura , Agrobacterium tumefaciens/genética , Conjugación Genética , Microscopía Electrónica de Transmisión , Mapas de Interacción de Proteínas
4.
Nature ; 508(7497): 550-553, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24670658

RESUMEN

Bacterial type IV secretion systems translocate virulence factors into eukaryotic cells, distribute genetic material between bacteria and have shown potential as a tool for the genetic modification of human cells. Given the complex choreography of the substrate through the secretion apparatus, the molecular mechanism of the type IV secretion system has proved difficult to dissect in the absence of structural data for the entire machinery. Here we use electron microscopy to reconstruct the type IV secretion system encoded by the Escherichia coli R388 conjugative plasmid. We show that eight proteins assemble in an intricate stoichiometric relationship to form an approximately 3 megadalton nanomachine that spans the entire cell envelope. The structure comprises an outer membrane-associated core complex connected by a central stalk to a substantial inner membrane complex that is dominated by a battery of 12 VirB4 ATPase subunits organized as side-by-side hexameric barrels. Our results show a secretion system with markedly different architecture, and consequently mechanism, to other known bacterial secretion systems.


Asunto(s)
Sistemas de Secreción Bacterianos , Escherichia coli/química , Escherichia coli/ultraestructura , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/ultraestructura , Sistemas de Secreción Bacterianos/genética , Membrana Celular/metabolismo , Escherichia coli/citología , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/aislamiento & purificación , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestructura , Microscopía Electrónica , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura
5.
Subcell Biochem ; 93: 53-81, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31939149

RESUMEN

Ribosomes are biological nanomachine that synthesise all proteins within a cell. It took decades to reveal the architecture of this essential cellular component. To understand the structure -function relationship of this nanomachine needed the utilisisation of different biochemical, biophysical and structural techniques. Structural studies combined with mutagenesis of the different ribosomal complexes comprising various RNAs and proteins enabled us to understand how this machine works inside a cell. Nowadays quite a number of ribosomal structures were published that confirmed biochemical studies on particular steps of protein synthesis by the ribosome . Four major steps were identified: initiation , elongation, termination and recycling. These steps lead us to the important question how the ribosome function can be regulated. Advances in technology for cryo electron microscopy: sample preparations, image recording, developments in algorithms for image analysis and processing significantly helped in revelation of structural details of the ribosome . We now have a library of ribosome structures from prokaryotes to eukaryotes that enable us to understand the complex mechanics of this nanomachine. As this structural library continues to grow, we gradually improve our understanding of this process and how it can be regulated and how the specific ribosomes can be stalled or activated, or completely disabled. This article provides a comprehensive overview of ribosomal structures that represent structural snapshots of the ribosome at its different functional states. Better understanding rises more particular questions that have to be addressed by determination structures of more complexes.Synopsis: Structural biology of the ribosome.


Asunto(s)
Biosíntesis de Proteínas , Ribosomas/química , Ribosomas/metabolismo , Microscopía por Crioelectrón , Ribosomas/ultraestructura
6.
Mol Microbiol ; 105(2): 273-293, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28452085

RESUMEN

Recent studies have shown that conjugation systems of Gram-negative bacteria are composed of distinct inner and outer membrane core complexes (IMCs and OMCCs, respectively). Here, we characterized the OMCC by focusing first on a cap domain that forms a channel across the outer membrane. Strikingly, the OMCC caps of the Escherichia coli pKM101 Tra and Agrobacterium tumefaciens VirB/VirD4 systems are completely dispensable for substrate transfer, but required for formation of conjugative pili. The pKM101 OMCC cap and extended pilus also are dispensable for activation of a Pseudomonas aeruginosa type VI secretion system (T6SS). Chimeric conjugation systems composed of the IMCpKM101 joined to OMCCs from the A. tumefaciens VirB/VirD4, E. coli R388 Trw, and Bordetella pertussis Ptl systems support conjugative DNA transfer in E. coli and trigger P. aeruginosa T6SS killing, but not pilus production. The A. tumefaciens VirB/VirD4 OMCC, solved by transmission electron microscopy, adopts a cage structure similar to the pKM101 OMCC. The findings establish that OMCCs are highly structurally and functionally conserved - but also intrinsically conformationally flexible - scaffolds for translocation channels. Furthermore, the OMCC cap and a pilus tip protein coregulate pilus extension but are not required for channel assembly or function.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Conjugación Genética/genética , Agrobacterium tumefaciens/genética , Proteínas de la Membrana Bacteriana Externa/fisiología , Proteínas Bacterianas/metabolismo , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Escherichia coli/genética , Fimbrias Bacterianas/metabolismo , Unión Proteica , Transporte de Proteínas/genética , Sistemas de Secreción Tipo IV/genética , Sistemas de Secreción Tipo IV/metabolismo , Factores de Virulencia/metabolismo
7.
Nucleic Acids Res ; 44(13): 6185-99, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27034469

RESUMEN

p53 tumor suppressor is a transcription factor that controls cell cycle and genetic integrity. In response to genotoxic stress p53 activates DNA repair, cell cycle arrest, apoptosis or senescence, which are initiated via p53 binding to its specific DNA response elements (RE). The consensus p53 DNA RE consists of two decameric palindromic half-site sequences. Crystallographic studies have demonstrated that two isolated p53 DNA-binding core domains interact with one half-site of the p53 DNA REs suggesting that one p53 tetramer is bound to one RE. However, our recent 3D cryo-EM studies showed that the full-length p53 tetramer is bound to only one half-site of RE.Here, we have used biochemical and electron microscopy (EM) methods to analyze DNA-binding of human and murine p53 tetramers to various p53 DNA REs. Our new results demonstrate that two p53 tetramers can interact sequence-specifically with one DNA RE at the same time. In particular, the EM structural analysis revealed that two p53 tetramers bind one DNA RE simultaneously with DNA positioned between them. These results demonstrate a mode different from that assumed previously for the p53-DNA interaction and suggest important biological implications on p53 activity as a transcriptional regulator of cellular response to stress.


Asunto(s)
Daño del ADN/genética , Proteínas de Unión al ADN/genética , Elementos de Respuesta/genética , Proteína p53 Supresora de Tumor/genética , Animales , Sitios de Unión/genética , Ciclo Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Secuencias Invertidas Repetidas/genética , Ratones , Conformación Proteica , Dominios Proteicos/genética , Multimerización de Proteína/genética , Estructura Terciaria de Proteína , Activación Transcripcional , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo
8.
Proc Natl Acad Sci U S A ; 112(22): 7009-14, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-25991862

RESUMEN

Many icosahedral viruses use a specialized portal vertex to control genome encapsidation and release from the viral capsid. In tailed bacteriophages, the portal system is connected to a tail structure that provides the pipeline for genome delivery to the host cell. We report the first, to our knowledge, subnanometer structures of the complete portal-phage tail interface that mimic the states before and after DNA release during phage infection. They uncover structural rearrangements associated with intimate protein-DNA interactions. The portal protein gp6 of bacteriophage SPP1 undergoes a concerted reorganization of the structural elements of its central channel during interaction with DNA. A network of protein-protein interactions primes consecutive binding of proteins gp15 and gp16 to extend and close the channel. This critical step that prevents genome leakage from the capsid is achieved by a previously unidentified allosteric mechanism: gp16 binding to two different regions of gp15 drives correct positioning and folding of an inner gp16 loop to interact with equivalent loops of the other gp16 subunits. Together, these loops build a plug that closes the channel. Gp16 then fastens the tail to yield the infectious virion. The gatekeeper system opens for viral genome exit at the beginning of infection but recloses afterward, suggesting a molecular diaphragm-like mechanism to control DNA efflux. The mechanisms described here, controlling the essential steps of phage genome movements during virus assembly and infection, are likely to be conserved among long-tailed phages, the largest group of viruses in the Biosphere.


Asunto(s)
Bacteriófagos/química , Genoma Viral/fisiología , Modelos Moleculares , Proteínas Virales/química , Proteínas de la Cola de los Virus/química , Ensamble de Virus/fisiología , Internalización del Virus , Bacteriófagos/ultraestructura , Microscopía por Crioelectrón , Genoma Viral/genética , Conformación Proteica , Proteínas Virales/metabolismo , Proteínas Virales/ultraestructura , Proteínas de la Cola de los Virus/metabolismo , Proteínas de la Cola de los Virus/ultraestructura
9.
EMBO J ; 32(8): 1195-204, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23511972

RESUMEN

Type IV secretion (T4S) systems are able to transport DNAs and/or proteins through the membranes of bacteria. They form large multiprotein complexes consisting of 12 proteins termed VirB1-11 and VirD4. VirB7, 9 and 10 assemble into a 1.07 MegaDalton membrane-spanning core complex (CC), around which all other components assemble. This complex is made of two parts, the O-layer inserted in the outer membrane and the I-layer inserted in the inner membrane. While the structure of the O-layer has been solved by X-ray crystallography, there is no detailed structural information on the I-layer. Using high-resolution cryo-electron microscopy and molecular modelling combined with biochemical approaches, we determined the I-layer structure and located its various components in the electron density. Our results provide new structural insights on the CC, from which the essential features of T4S system mechanisms can be derived.


Asunto(s)
Agrobacterium tumefaciens/química , Sistemas de Secreción Bacterianos , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/ultraestructura , Complejos Multiproteicos/química , Complejos Multiproteicos/ultraestructura , Microscopía por Crioelectrón , Modelos Moleculares , Conformación Proteica
10.
Nucleic Acids Res ; 43(17): 8551-63, 2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26240379

RESUMEN

Hexameric helicases are processive DNA unwinding machines but how they engage with a replication fork during unwinding is unknown. Using electron microscopy and single particle analysis we determined structures of the intact hexameric helicase E1 from papillomavirus and two complexes of E1 bound to a DNA replication fork end-labelled with protein tags. By labelling a DNA replication fork with streptavidin (dsDNA end) and Fab (5' ssDNA) we located the positions of these labels on the helicase surface, showing that at least 10 bp of dsDNA enter the E1 helicase via a side tunnel. In the currently accepted 'steric exclusion' model for dsDNA unwinding, the active 3' ssDNA strand is pulled through a central tunnel of the helicase motor domain as the dsDNA strands are wedged apart outside the protein assembly. Our structural observations together with nuclease footprinting assays indicate otherwise: strand separation is taking place inside E1 in a chamber above the helicase domain and the 5' passive ssDNA strands exits the assembly through a separate tunnel opposite to the dsDNA entry point. Our data therefore suggest an alternative to the current general model for DNA unwinding by hexameric helicases.


Asunto(s)
ADN Helicasas/química , ADN/química , ADN/metabolismo , ADN Helicasas/metabolismo , ADN Helicasas/ultraestructura , Replicación del ADN , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/ultraestructura , Modelos Moleculares , Papillomaviridae/enzimología , Estructura Terciaria de Proteína
11.
J Biol Chem ; 290(12): 7973-9, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25648893

RESUMEN

ATP-dependent DNA unwinding activity has been demonstrated for recombinant archaeal homohexameric minichromosome maintenance (MCM) complexes and their yeast heterohexameric counterparts, but in higher eukaryotes such as Drosophila, MCM-associated DNA helicase activity has been observed only in the context of a co-purified Cdc45-MCM-GINS complex. Here, we describe the production of the recombinant human MCM (hMCM) complex in Escherichia coli. This protein displays ATP hydrolysis activity and is capable of unwinding duplex DNA. Using single-particle asymmetric EM reconstruction, we demonstrate that recombinant hMCM forms a hexamer that undergoes a conformational change when bound to DNA. Recombinant hMCM produced without post-translational modifications is functional in vitro and provides an important tool for biochemical reconstitution of the human replicative helicase.


Asunto(s)
Cromosomas Humanos , ADN/química , Proteínas de Mantenimiento de Minicromosoma/química , Secuencia de Bases , Humanos , Datos de Secuencia Molecular , Conformación Proteica
12.
J Biol Chem ; 290(6): 3836-49, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25525268

RESUMEN

The majority of known bacteriophages have long tails that serve for bacterial target recognition and viral DNA delivery into the host. These structures form a tube from the viral capsid to the bacterial cell. The tube is formed primarily by a helical array of tail tube protein (TTP) subunits. In phages with a contractile tail, the TTP tube is surrounded by a sheath structure. Here, we report the first evidence that a phage TTP, gp17.1 of siphophage SPP1, self-assembles into long tubes in the absence of other viral proteins. gp17.1 does not exhibit a stable globular structure when monomeric in solution, even if it was confidently predicted to adopt the ß-sandwich fold of phage λ TTP. However, Fourier transform infrared and nuclear magnetic resonance spectroscopy analyses showed that its ß-sheet content increases significantly during tube assembly, suggesting that gp17.1 acquires a stable ß-sandwich fold only after self-assembly. EM analyses revealed that the tube is formed by hexameric rings stacked helicoidally with the same organization and helical parameters found for the tail of SPP1 virions. These parameters were used to build a pseudo-atomic model of the TTP tube. The large loop spanning residues 40-56 is located on the inner surface of the tube, at the interface between adjacent monomers and hexamers. In line with our structural predictions, deletion of this loop hinders gp17.1 tube assembly in vitro and interferes with SPP1 tail assembly during phage particle morphogenesis in bacteria.


Asunto(s)
Pliegue de Proteína , Proteínas Virales/química , Secuencia de Aminoácidos , Bacteriófagos/química , Datos de Secuencia Molecular , Estructura Terciaria de Proteína
13.
Nature ; 468(7322): 447-51, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21037563

RESUMEN

Natural killer cells and cytotoxic T lymphocytes accomplish the critically important function of killing virus-infected and neoplastic cells. They do this by releasing the pore-forming protein perforin and granzyme proteases from cytoplasmic granules into the cleft formed between the abutting killer and target cell membranes. Perforin, a 67-kilodalton multidomain protein, oligomerizes to form pores that deliver the pro-apoptopic granzymes into the cytosol of the target cell. The importance of perforin is highlighted by the fatal consequences of congenital perforin deficiency, with more than 50 different perforin mutations linked to familial haemophagocytic lymphohistiocytosis (type 2 FHL). Here we elucidate the mechanism of perforin pore formation by determining the X-ray crystal structure of monomeric murine perforin, together with a cryo-electron microscopy reconstruction of the entire perforin pore. Perforin is a thin 'key-shaped' molecule, comprising an amino-terminal membrane attack complex perforin-like (MACPF)/cholesterol dependent cytolysin (CDC) domain followed by an epidermal growth factor (EGF) domain that, together with the extreme carboxy-terminal sequence, forms a central shelf-like structure. A C-terminal C2 domain mediates initial, Ca(2+)-dependent membrane binding. Most unexpectedly, however, electron microscopy reveals that the orientation of the perforin MACPF domain in the pore is inside-out relative to the subunit arrangement in CDCs. These data reveal remarkable flexibility in the mechanism of action of the conserved MACPF/CDC fold and provide new insights into how related immune defence molecules such as complement proteins assemble into pores.


Asunto(s)
Membrana Celular/metabolismo , Linfocitos/metabolismo , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Animales , Colesterol/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Factor de Crecimiento Epidérmico/química , Granzimas/metabolismo , Humanos , Ratones , Modelos Moleculares , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/ultraestructura , Estructura Terciaria de Proteína
14.
Proc Natl Acad Sci U S A ; 110(14): 5468-73, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23513222

RESUMEN

The cross-ß amyloid form of peptides and proteins represents an archetypal and widely accessible structure consisting of ordered arrays of ß-sheet filaments. These complex aggregates have remarkable chemical and physical properties, and the conversion of normally soluble functional forms of proteins into amyloid structures is linked to many debilitating human diseases, including several common forms of age-related dementia. Despite their importance, however, cross-ß amyloid fibrils have proved to be recalcitrant to detailed structural analysis. By combining structural constraints from a series of experimental techniques spanning five orders of magnitude in length scale--including magic angle spinning nuclear magnetic resonance spectroscopy, X-ray fiber diffraction, cryoelectron microscopy, scanning transmission electron microscopy, and atomic force microscopy--we report the atomic-resolution (0.5 Å) structures of three amyloid polymorphs formed by an 11-residue peptide. These structures reveal the details of the packing interactions by which the constituent ß-strands are assembled hierarchically into protofilaments, filaments, and mature fibrils.


Asunto(s)
Amiloide/química , Amiloide/ultraestructura , Modelos Moleculares , Estructura Secundaria de Proteína , Microscopía por Crioelectrón , Espectroscopía de Resonancia Magnética/métodos , Microscopía Electrónica de Transmisión de Rastreo , Difracción de Rayos X
15.
Proc Natl Acad Sci U S A ; 109(28): 11348-53, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22745169

RESUMEN

Type IV secretion (T4S) systems mediate the transfer of proteins and DNA across the cell envelope of bacteria. These systems play important roles in bacterial pathogenesis and in horizontal transfer of antibiotic resistance. The VirB4 ATPase of the T4S system is essential for both the assembly of the system and substrate transfer. In this article, we present the crystal structure of the C-terminal domain of Thermoanaerobacter pseudethanolicus VirB4. This structure is strikingly similar to that of another T4S ATPase, VirD4, a protein that shares only 12% sequence identity with VirB4. The VirB4 domain purifies as a monomer, but the full-length protein is observed in a monomer-dimer equilibrium, even in the presence of nucleotides and DNAs. We also report the negative stain electron microscopy structure of the core complex of the T4S system of the Escherichia coli pKM101 plasmid, with VirB4 bound. In this structure, VirB4 is also monomeric and bound through its N-terminal domain to the core's VirB9 protein. Remarkably, VirB4 is observed bound to the side of the complex where it is ideally placed to play its known regulatory role in substrate transfer.


Asunto(s)
Adenosina Trifosfatasas/química , Proteínas Bacterianas/química , Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Sitios de Unión , Cristalización , Cristalografía por Rayos X/métodos , ADN Bacteriano/genética , Escherichia coli/metabolismo , Sustancias Macromoleculares/metabolismo , Magnesio/química , Espectrometría de Masas/métodos , Microscopía Electrónica/métodos , Modelos Biológicos , Nucleótidos/química , Plásmidos , Unión Proteica , Conformación Proteica , Factores de Virulencia/genética
16.
Proc Natl Acad Sci U S A ; 109(50): 20455-60, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23184970

RESUMEN

Protein misfolding and aggregation cause serious degenerative conditions such as Alzheimer's, Parkinson, and prion diseases. Damage to membranes is thought to be one of the mechanisms underlying cellular toxicity of a range of amyloid assemblies. Previous studies have indicated that amyloid fibrils can cause membrane leakage and elicit cellular damage, and these effects are enhanced by fragmentation of the fibrils. Here we report direct 3D visualization of membrane damage by specific interactions of a lipid bilayer with amyloid-like fibrils formed in vitro from ß(2)-microglobulin (ß(2)m). Using cryoelectron tomography, we demonstrate that fragmented ß(2)m amyloid fibrils interact strongly with liposomes and cause distortions to the membranes. The normally spherical liposomes form pointed teardrop-like shapes with the fibril ends seen in proximity to the pointed regions on the membranes. Moreover, the tomograms indicated that the fibrils extract lipid from the membranes at these points of distortion by removal or blebbing of the outer membrane leaflet. Tiny (15-25 nm) vesicles, presumably formed from the extracted lipids, were observed to be decorating the fibrils. The findings highlight a potential role of fibrils, and particularly fibril ends, in amyloid pathology, and report a previously undescribed class of lipid-protein interactions in membrane remodelling.


Asunto(s)
Amiloide/química , Amiloide/ultraestructura , Animales , Fenómenos Biofísicos , Microscopía por Crioelectrón/métodos , Tomografía con Microscopio Electrónico/métodos , Humanos , Liposomas/química , Liposomas/ultraestructura , Membranas/química , Membranas/ultraestructura , Microscopía Fluorescente , Multimerización de Proteína , Microglobulina beta-2/química , Microglobulina beta-2/ultraestructura
17.
J Virol ; 86(12): 6768-77, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22514336

RESUMEN

The structure of the bacteriophage SPP1 capsid was determined at subnanometer resolution by cryo-electron microscopy and single-particle analysis. The icosahedral capsid is composed of the major capsid protein gp13 and the auxiliary protein gp12, which are organized in a T=7 lattice. DNA is arranged in layers with a distance of ~24.5 Å. gp12 forms spikes that are anchored at the center of gp13 hexamers. In a gp12-deficient mutant, the centers of hexamers are closed by loops of gp13 coming together to protect the SPP1 genome from the outside environment. The HK97-like fold was used to build a pseudoatomic model of gp13. Its structural organization remains unchanged upon tail binding and following DNA release. gp13 exhibits enhanced thermostability in the DNA-filled capsid. A remarkable convergence between the thermostability of the capsid and those of the other virion components was found, revealing that the overall architecture of the SPP1 infectious particle coevolved toward high robustness.


Asunto(s)
Bacteriófagos/fisiología , Cápside/química , Cápside/metabolismo , Ensamble de Virus , Bacteriófagos/química , Bacteriófagos/genética , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Microscopía por Crioelectrón , Modelos Moleculares , Estabilidad Proteica
18.
Nucleic Acids Res ; 39(20): 8960-71, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21764777

RESUMEN

The p53 tumour suppressor is a transcriptional activator that controls cell fate in response to various stresses. p53 can initiate cell cycle arrest, senescence and/or apoptosis via transactivation of p53 target genes, thus preventing cancer onset. Mutations that impair p53 usually occur in the core domain and negate the p53 sequence-specific DNA binding. Moreover, these mutations exhibit a dominant negative effect on the remaining wild-type p53. Here, we report the cryo electron microscopy structure of the full-length p53 tetramer bound to a DNA-encoding transcription factor response element (RE) at a resolution of 21 A. While two core domains from both dimers of the p53 tetramer interact with DNA within the complex, the other two core domains remain available for binding another DNA site. This finding helps to explain the dominant negative effect of p53 mutants based on the fact that p53 dimers are formed co-translationally before the whole tetramer assembles; therefore, a single mutant dimer would prevent the p53 tetramer from binding DNA. The structure indicates that the Achilles' heel of p53 is in its dimer-of-dimers organization, thus the tetramer activity can be negated by mutation in only one allele followed by tumourigenesis.


Asunto(s)
ADN/química , Mutación , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Animales , Microscopía por Crioelectrón , ADN/ultraestructura , Ratones , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , Estructura Terciaria de Proteína , Elementos de Respuesta , Proteína p53 Supresora de Tumor/ultraestructura
19.
Nat Commun ; 14(1): 3630, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37336895

RESUMEN

DNA can be folded into rationally designed, unique, and functional materials. To fully realise the potential of these DNA materials, a fundamental understanding of their structure and dynamics is necessary, both in simple solvents as well as more complex and diverse anisotropic environments. Here we analyse an archetypal six-duplex DNA nanoarchitecture with single-particle cryo-electron microscopy and molecular dynamics simulations in solvents of tunable ionic strength and within the anisotropic environment of biological membranes. Outside lipid bilayers, the six-duplex bundle lacks the designed symmetrical barrel-type architecture. Rather, duplexes are arranged in non-hexagonal fashion and are disorted to form a wider, less elongated structure. Insertion into lipid membranes, however, restores the anticipated barrel shape due to lateral duplex compression by the bilayer. The salt concentration has a drastic impact on the stability of the inserted barrel-shaped DNA nanopore given the tunable electrostatic repulsion between the negatively charged duplexes. By synergistically combining experiments and simulations, we increase fundamental understanding into the environment-dependent structural dynamics of a widely used nanoarchitecture. This insight will pave the way for future engineering and biosensing applications.


Asunto(s)
Membrana Dobles de Lípidos , Simulación de Dinámica Molecular , Microscopía por Crioelectrón , Membrana Celular/química , Membrana Dobles de Lípidos/química , ADN/química , Solventes
20.
Adv Exp Med Biol ; 726: 585-600, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22297531

RESUMEN

Tailed bacteriophages use a portal system for genome entry and exit from viral capsids. Here, we review the mechanisms how these movements are controlled by the genome gatekeeper that assembles at the portal structure. Phage DNA is packaged at high pressure inside the viral capsid by a powerful motor. The viral genome is translocated through the central channel of the portal protein found at a single vertex of the capsid. Packaging is normally terminated by endonucleolytic cleavage of the substrate DNA followed by disassembly of the packaging motor and closure of the portal system, preventing leakage of the viral genome. This can be achieved either by conformational changes in the portal protein or by sequential addition of proteins that extend the portal channel (adaptors) and physically close it preventing DNA exit (stoppers). The resulting connector structure provides the interface for assembly of short tails (podoviruses) or for attachment of preformed long tails (siphoviruses and myoviruses). The connector maintains the viral DNA correctly positioned for ejection that is triggered by interaction of the phage particle with bacterial receptors. Recent exciting advances are providing new molecular insights on the mechanisms that ensure precise coordination of these critical steps required both for stable viral genome packaging and for its efficient release to initiate infection.


Asunto(s)
Bacteriófagos , Cápside , Empaquetamiento del ADN , Genoma Viral , Ensamble de Virus/genética , Bacteriófagos/genética , Bacteriófagos/metabolismo , Bacteriófagos/ultraestructura , Cápside/metabolismo , Cápside/ultraestructura , ADN Viral/química , ADN Viral/metabolismo , Modelos Moleculares , Estructura Cuaternaria de Proteína , Proteínas Virales/química , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA