Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 598(7881): 521-525, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34526719

RESUMEN

Hepatitis C virus (HCV) infection is a causal agent of chronic liver disease, cirrhosis and hepatocellular carcinoma in humans, and afflicts more than 70 million people worldwide. The HCV envelope glycoproteins E1 and E2 are responsible for the binding of the virus to the host cell, but the exact entry process remains undetermined1. The majority of broadly neutralizing antibodies block interaction between HCV E2 and the large extracellular loop (LEL) of the cellular receptor CD81 (CD81-LEL)2. Here we show that low pH enhances the binding of CD81-LEL to E2, and we determine the crystal structure of E2 in complex with an antigen-binding fragment (2A12) and CD81-LEL (E2-2A12-CD81-LEL); E2 in complex with 2A12 (E2-2A12); and CD81-LEL alone. After binding CD81, residues 418-422 in E2 are displaced, which allows for the extension of an internal loop consisting of residues 520-539. Docking of the E2-CD81-LEL complex onto a membrane-embedded, full-length CD81 places the residues Tyr529 and Trp531 of E2 proximal to the membrane. Liposome flotation assays show that low pH and CD81-LEL increase the interaction of E2 with membranes, whereas structure-based mutants of Tyr529, Trp531 and Ile422 in the amino terminus of E2 abolish membrane binding. These data support a model in which acidification and receptor binding result in a conformational change in E2 in preparation for membrane fusion.


Asunto(s)
Hepacivirus/metabolismo , Unión Proteica , Receptores Virales/química , Receptores Virales/metabolismo , Tetraspanina 28/química , Tetraspanina 28/metabolismo , Internalización del Virus , Animales , Anticuerpos Neutralizantes/inmunología , Membrana Celular/química , Membrana Celular/metabolismo , Células HEK293 , Hepacivirus/química , Hepacivirus/genética , Humanos , Leontopithecus , Fusión de Membrana , Modelos Moleculares , Receptores Virales/inmunología , Tetraspanina 28/inmunología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo
2.
J Virol ; 98(1): e0084923, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38174935

RESUMEN

Hepatitis C virus (HCV) is a member of the Flaviviridae family; however, unlike other family members, the HCV virion has an unusually high lipid content. HCV has two envelope glycoproteins, E1 and E2. E2 contributes to receptor binding, cell membrane attachment, and immune evasion. In contrast, the functions of E1 are poorly characterized due, in part, to challenges in producing the protein. This manuscript describes the expression and purification of a soluble E1 ectodomain (eE1) that is recognized by conformational, human monoclonal antibodies. eE1 forms a complex with apolipoproteins AI and AII, cholesterol, and phospholipids by recruiting high-density lipoprotein (HDL) from the extracellular media. We show that HDL binding is a function specific to eE1 and HDL hinders recognition of E1 by a neutralizing monoclonal antibody. Either low-density lipoprotein or HDL increases the production and infectivity of cell culture-produced HCV, but E1 preferentially selects HDL, influencing both viral life cycle and antibody evasion.IMPORTANCEHepatitis C virus (HCV) infection is a significant burden on human health, but vaccine candidates have yet to provide broad protection against this infection. We have developed a method to produce high quantities of soluble E1 or E2, the viral proteins located on the surface of HCV. HCV has an unusually high lipid content due to the recruitment of apolipoproteins. We found that E1 (and not E2) preferentially recruits host high-density lipoprotein (HDL) extracellularly. This recruitment of HDL by E1 prevents binding of E1 by a neutralizing antibody and furthermore prevents antibody-mediated neutralization of the virus. By comparison, low-density lipoprotein does not protect the virus from antibody-mediated neutralization. Our findings provide mechanistic insight into apolipoprotein recruitment, which may be critical for vaccine development.


Asunto(s)
Hepacivirus , Hepatitis C , Evasión Inmune , Lipoproteínas HDL , Proteínas del Envoltorio Viral , Humanos , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Apolipoproteínas/metabolismo , Hepacivirus/patogenicidad , Hepatitis C/inmunología , Hepatitis C/virología , Anticuerpos contra la Hepatitis C/inmunología , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Células HEK293
3.
PLoS Pathog ; 16(2): e1008314, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32069326

RESUMEN

Staphylococcus aureus is a common cause of infections in humans. The emergence of virulent, antibiotic-resistant strains of S. aureus is a significant public health concern. Most virulence and resistance factors in S. aureus are encoded by mobile genetic elements, and transduction by bacteriophages represents the main mechanism for horizontal gene transfer. The baseplate is a specialized structure at the tip of bacteriophage tails that plays key roles in host recognition, cell wall penetration, and DNA ejection. We have used high-resolution cryo-electron microscopy to determine the structure of the S. aureus bacteriophage 80α baseplate at 3.75 Å resolution, allowing atomic models to be built for most of the major tail and baseplate proteins, including two tail fibers, the receptor binding protein, and part of the tape measure protein. Our structure provides a structural basis for understanding host recognition, cell wall penetration and DNA ejection in viruses infecting Gram-positive bacteria. Comparison to other phages demonstrates the modular design of baseplate proteins, and the adaptations to the host that take place during the evolution of staphylococci and other pathogens.


Asunto(s)
Bacteriófagos/metabolismo , Bacteriófagos/ultraestructura , Staphylococcus aureus/genética , Bacteriófagos/genética , Microscopía por Crioelectrón/métodos , Transferencia de Gen Horizontal/genética , Humanos , Modelos Moleculares , Unión Proteica/genética , Conformación Proteica , Staphylococcus aureus/ultraestructura , Staphylococcus aureus/virología , Virión/genética
4.
J Biol Chem ; 292(40): 16760-16772, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28842495

RESUMEN

Hepatitis B virus (HBV) infection afflicts millions worldwide, causing cirrhosis and liver cancer. HBV e-antigen (HBeAg), a clinical marker for disease severity, is a soluble variant of the viral capsid protein. HBeAg is not required for viral replication but is implicated in establishing immune tolerance and chronic infection. The structure of recombinant e-antigen (rHBeAg) was recently determined, yet to date, the exact nature and quantitation of HBeAg still remain uncertain. Here, to further characterize HBeAg, we used phage display to produce a panel of chimeric rabbit/human monoclonal antibody fragments (both Fab and scFv) against rHBeAg. Several of the Fab/scFv, expressed in Escherichia coli, had unprecedentedly high binding affinities (Kd ∼10-12 m) and high specificity. We used Fab/scFv in the context of an enzyme-linked immunosorbent assay (ELISA) for HBeAg quantification, which we compared with commercially available kits and verified with seroconversion panels, the WHO HBeAg standard, rHBeAg, and patient plasma samples. We found that the specificity and sensitivity are superior to those of existing commercial assays. To identify potential fine differences between rHBeAg and HBeAg, we used these Fabs in microscale immunoaffinity chromatography to purify HBeAg from individual patient plasmas. Western blotting and MS results indicated that rHBeAg and HBeAg are essentially structurally identical, although HBeAg from different patients exhibits minor carboxyl-terminal heterogeneity. We discuss several potential applications for the humanized Fab/scFv.


Asunto(s)
Anticuerpos contra la Hepatitis B/inmunología , Antígenos e de la Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Hepatitis B/inmunología , Proteínas Recombinantes de Fusión/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática/métodos , Hepatitis B/sangre , Hepatitis B/tratamiento farmacológico , Anticuerpos contra la Hepatitis B/química , Anticuerpos contra la Hepatitis B/genética , Anticuerpos contra la Hepatitis B/uso terapéutico , Antígenos e de la Hepatitis B/sangre , Antígenos e de la Hepatitis B/química , Virus de la Hepatitis B/química , Humanos , Conejos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/uso terapéutico
5.
Biochemistry ; 56(14): 2061-2070, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28349680

RESUMEN

Z-disc-associated, alternatively spliced, PDZ motif-containing protein (ZASP) is a principal component of the sarcomere. The three prevalent isoforms of ZASP in skeletal muscle are generated by alternative splicing of exons 9 and 10. The long isoforms, either having (ZASP-L) or lacking exon 10 (ZASP-LΔex10), include an N-terminal PDZ domain, an actin-binding region (ABR) with a conserved motif (ZM), and three C-terminal LIM domains. The short isoform (ZASP-S) lacks the LIM domains. Mutations, A147T and A165V, within the ZM of ZASP-LΔex10 cause myofibrillar myopathy, but the mechanism is unknown. We have prepared these proteins, their ABR, and the respective mutant variants in recombinant form, characterized them biophysically, and analyzed their actin-binding properties by surface plasmon resonance and electron microscopy. All the proteins were physically homogeneous and monomeric and had circular dichroic spectra consistent with partially folded conformations. Comparison of the NMR HSQC spectra of ZASP-S and the PDZ domain showed that the ABR is unstructured. ZASP-S and its mutant variants and ZASP-LΔex10 all bound to immobilized G-actin with high affinity (Kd ≈ 10-8 to 10-9 M). Constructs of the isolated actin-binding region missing exon 10 (ABRΔ10) bound with lower affinity (Kd ≈ 10-7 M), but those retaining exon 10 (ABR+10) did so only weakly (Kd ≈ 10-5 M). ZASP-S, and the ABRΔ10, also induced F-actin and array formation, even in conditions of low ionic strength and in the absence of KCl and Mg2+ ions. Interestingly, the ZM mutations A147T and A165V did not affect any of the results described above.


Asunto(s)
Actinas/química , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas con Dominio LIM/química , Actinas/genética , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme Alternativo , Sitios de Unión , Escherichia coli/genética , Escherichia coli/metabolismo , Exones , Expresión Génica , Humanos , Intrones , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Mutación , Concentración Osmolar , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sarcómeros/fisiología , Relación Estructura-Actividad
6.
J Biol Chem ; 291(5): 2310-8, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26644467

RESUMEN

Parkinson disease and other progressive neurodegenerative conditions are characterized by the intracerebral presence of Lewy bodies, containing amyloid fibrils of α-synuclein. We used cryo-electron microscopy and scanning transmission electron microscopy (STEM) to study in vitro-assembled fibrils. These fibrils are highly polymorphic. Focusing on twisting fibrils with an inter-crossover spacing of 77 nm, our reconstructions showed them to consist of paired protofibrils. STEM mass per length data gave one subunit per 0.47 nm axial rise per protofibril, consistent with a superpleated ß-structure. The STEM images show two thread-like densities running along each of these fibrils, which we interpret as ladders of metal ions. These threads confirmed the two-protofibril architecture of the 77-nm twisting fibrils and allowed us to identify this morphotype in STEM micrographs. Some other, but not all, fibril morphotypes also exhibit dense threads, implying that they also present a putative metal binding site. We propose a molecular model for the protofibril and suggest that polymorphic variant fibrils have different numbers of protofibrils that are associated differently.


Asunto(s)
Amiloide/química , alfa-Sinucleína/química , Secuencia de Aminoácidos , Sitios de Unión , Microscopía por Crioelectrón , Humanos , Procesamiento de Imagen Asistido por Computador , Iones , Cuerpos de Lewy/metabolismo , Microscopía Electrónica de Transmisión de Rastreo , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Homología de Secuencia de Aminoácido
7.
Elife ; 122023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36947136

RESUMEN

A domain in the ORF1 polyprotein of the hepatitis E virus that was previously thought to be a protease is actually a zinc-binding domain.


Asunto(s)
Virus de la Hepatitis E , Virus de la Hepatitis E/genética , Péptido Hidrolasas , Poliproteínas , Replicación Viral
8.
Nat Commun ; 14(1): 433, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-36702826

RESUMEN

Hepatitis C virus (HCV) uses a hybrid entry mechanism. Current structural data suggest that upon exposure to low pH and Cluster of Differentiation 81 (CD81), the amino terminus of envelope glycoprotein E2 becomes ordered and releases an internal loop with two invariant aromatic residues into the host membrane. Here, we present the structure of an amino-terminally truncated E2 with the membrane binding loop in a bent conformation and the aromatic side chains sequestered. Comparison with three previously reported E2 structures with the same Fab indicates that this internal loop is flexible, and that local context influences the exposure of hydrophobic residues. Biochemical assays show that the amino-terminally truncated E2 lacks the baseline membrane-binding capacity of the E2 ectodomain. Thus, the amino terminal region is a critical determinant for both CD81 and membrane interaction. These results provide new insights into the HCV entry mechanism.


Asunto(s)
Hepacivirus , Hepatitis C , Humanos , Hepacivirus/metabolismo , Unión Proteica , Proteínas del Envoltorio Viral/metabolismo , Tetraspanina 28/química , Tetraspanina 28/metabolismo
9.
J Struct Biol ; 178(3): 215-24, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22508104

RESUMEN

Bacteriophage P4 is dependent on structural proteins supplied by a helper phage, P2, to assemble infectious virions. Bacteriophage P2 normally forms an icosahedral capsid with T=7 symmetry from the gpN capsid protein, the gpO scaffolding protein and the gpQ portal protein. In the presence of P4, however, the same structural proteins are assembled into a smaller capsid with T=4 symmetry. This size determination is effected by the P4-encoded protein Sid, which forms an external scaffold around the small P4 procapsids. Size responsiveness (sir) mutants in gpN fail to assemble small capsids even in the presence of Sid. We have produced large and small procapsids by co-expression of gpN with gpO and Sid, respectively, and applied cryo-electron microscopy and three-dimensional reconstruction methods to visualize these procapsids. gpN has an HK97-like fold and interacts with Sid in an exposed loop where the sir mutations are clustered. The T=7 lattice of P2 has dextro handedness, unlike the laevo lattices of other phages with this fold observed so far.


Asunto(s)
Bacteriófago P2/química , Bacteriófago P2/ultraestructura , Cápside/química , Cápside/diagnóstico por imagen , Myoviridae/química , Myoviridae/ultraestructura , Bacteriófago P2/genética , Microscopía por Crioelectrón , Modelos Biológicos , Mutación , Myoviridae/genética , Estructura Secundaria de Proteína , Ultrasonografía
10.
Artículo en Inglés | MEDLINE | ID: mdl-31501263

RESUMEN

Hepatitis C virus (HCV) represents an important and growing public health problem, chronically infecting an estimated 70 million people worldwide. This blood-borne pathogen is generating a new wave of infections in the United States, associated with increasing intravenous drug use over the last decade. In most cases, HCV establishes a chronic infection, sometimes causing cirrhosis, end-stage liver disease, and hepatocellular carcinoma. Although a curative therapy exists, it is extremely expensive and provides no barrier to reinfection; therefore, a vaccine is urgently needed. The virion is asymmetric and heterogeneous with the buoyancy and protein content similar to low-density lipoparticles. Core protein is unstructured, and of the two envelope glycoproteins, E1 and E2, the function of E1 remains enigmatic. E2 is responsible for specifically binding host receptors CD81 and scavenger receptor class B type I (SR-BI). This review will focus on structural progress on HCV virion, core protein, envelope glycoproteins, and specific host receptors.


Asunto(s)
Hepacivirus/química , Proteínas del Envoltorio Viral/química , Anticuerpos Neutralizantes/química , Cristalografía por Rayos X , Hepacivirus/inmunología , Hepacivirus/metabolismo , Humanos , Estructura Terciaria de Proteína , Tetraspanina 28/química , Proteínas del Envoltorio Viral/inmunología , Vacunas contra Hepatitis Viral/química , Vacunas contra Hepatitis Viral/inmunología , Internalización del Virus
11.
J Cell Biol ; 218(3): 1027-1038, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30630865

RESUMEN

Mutations in the retinal protein retinoschisin (RS1) cause progressive loss of vision in young males, a form of macular degeneration called X-linked retinoschisis (XLRS). We previously solved the structure of RS1, a 16-mer composed of paired back-to-back octameric rings. Here, we show by cryo-electron microscopy that RS1 16-mers can assemble into extensive branched networks. We classified the different configurations, finding four types of interaction between the RS1 molecules. The predominant configuration is a linear strand with a wavy appearance. Three less frequent types constitute the branch points of the network. In all cases, the "spikes" around the periphery of the double rings are involved in these interactions. In the linear strand, a loop (usually referred to as spike 1) occurs on both sides of the interface between neighboring molecules. Mutations in this loop suppress secretion, indicating the possibility of intracellular higher-order assembly. These observations suggest that branched networks of RS1 may play a stabilizing role in maintaining the integrity of the retina.


Asunto(s)
Microscopía por Crioelectrón , Proteínas del Ojo/metabolismo , Mutación , Retina/metabolismo , Retina/ultraestructura , Retinosquisis/metabolismo , Retinosquisis/patología , Proteínas del Ojo/genética , Células HEK293 , Humanos , Masculino , Estructura Secundaria de Proteína , Retinosquisis/genética
12.
Structure ; 26(9): 1187-1195.e4, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30017564

RESUMEN

HIV-1 Rev protein mediates nuclear export of unspliced and partially spliced viral RNAs for production of viral genomes and structural proteins. Rev assembles on a 351-nt Rev response element (RRE) within viral transcripts and recruits host export machinery. Small (<40-nt) RNA aptamers that compete with the RRE for Rev binding inhibit HIV-1 viral replication. We determined the X-ray crystal structure of a potential anti-HIV-1 aptamer that binds Rev with high affinity (Kd = 5.9 nM). The aptamer is structurally similar to the RRE high-affinity site but forms additional contacts with Rev unique to its sequence. Exposed bases of the aptamer interleave with the guanidinium groups of two arginines of Rev, forming stacking interactions and hydrogen bonds. The aptamer also obstructs an oligomerization interface of Rev, blocking Rev self-assembly. We propose that this aptamer can inhibit HIV-1 replication by interfering with Rev-RRE, Rev-Rev, and possibly Rev-host protein interactions.


Asunto(s)
Aptámeros de Nucleótidos/química , VIH-1/fisiología , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/química , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Aptámeros de Nucleótidos/farmacología , Arginina/metabolismo , Sitios de Unión , Cristalografía por Rayos X , VIH-1/efectos de los fármacos , VIH-1/metabolismo , Modelos Moleculares , Unión Proteica/efectos de los fármacos , Elementos de Respuesta , Replicación Viral/efectos de los fármacos , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética
13.
Structure ; 26(10): 1314-1326.e4, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30100358

RESUMEN

Hepatitis B virus (HBV) is the leading cause of liver disease worldwide. While an adequate vaccine is available, current treatment options are limited, not highly effective, and associated with adverse effects, encouraging the development of alternative therapeutics. The HBV core gene encodes two different proteins: core, which forms the viral nucleocapsid, and pre-core, which serves as an immune modulator with multiple points of action. The two proteins mostly have the same sequence, although they differ at their N and C termini and in their dimeric arrangements. Previously, we engineered two human-framework antibody fragments (Fab/scFv) with nano- to picomolar affinities for both proteins. Here, by means of X-ray crystallography, analytical ultracentrifugation, and electron microscopy, we demonstrate that the antibodies have non-overlapping epitopes and effectively block biologically important assemblies of both proteins. These properties, together with the anticipated high tolerability and long half-lives of the antibodies, make them promising therapeutics.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos del Núcleo de la Hepatitis B/química , Antígenos e de la Hepatitis B/química , Virus de la Hepatitis B/metabolismo , Animales , Anticuerpos Monoclonales/química , Anticuerpos Antivirales , Sitios de Unión , Cristalografía por Rayos X , Antígenos del Núcleo de la Hepatitis B/metabolismo , Antígenos e de la Hepatitis B/metabolismo , Virus de la Hepatitis B/química , Humanos , Microscopía Electrónica , Modelos Moleculares , Unión Proteica , Estructura Cuaternaria de Proteína , Ultracentrifugación
14.
Elife ; 62017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28984245

RESUMEN

Staphylococcus aureus pathogenicity islands (SaPIs), such as SaPI1, exploit specific helper bacteriophages, like 80α, for their high frequency mobilization, a process termed 'molecular piracy'. SaPI1 redirects the helper's assembly pathway to form small capsids that can only accommodate the smaller SaPI1 genome, but not a complete phage genome. SaPI1 encodes two proteins, CpmA and CpmB, that are responsible for this size redirection. We have determined the structures of the 80α and SaPI1 procapsids to near-atomic resolution by cryo-electron microscopy, and show that CpmB competes with the 80α scaffolding protein (SP) for a binding site on the capsid protein (CP), and works by altering the angle between capsomers. We probed these interactions genetically and identified second-site suppressors of lethal mutations in SP. Our structures show, for the first time, the detailed interactions between SP and CP in a bacteriophage, providing unique insights into macromolecular assembly processes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteriófagos/metabolismo , Cápside/metabolismo , Islas Genómicas , Staphylococcus aureus/genética , Staphylococcus aureus/virología , Proteínas Virales/metabolismo , Ensamble de Virus , Proteínas Bacterianas/genética , Bacteriófagos/ultraestructura , Cápside/ultraestructura , Microscopía por Crioelectrón , Mapeo de Interacción de Proteínas , Proteínas Virales/genética
15.
Viruses ; 9(12)2017 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-29258203

RESUMEN

In the tailed bacteriophages, DNA is packaged into spherical procapsids, leading to expansion into angular, thin-walled mature capsids. In many cases, this maturation is accompanied by cleavage of the major capsid protein (CP) and other capsid-associated proteins, including the scaffolding protein (SP) that serves as a chaperone for the assembly process. Staphylococcus aureus bacteriophage 80α is capable of high frequency mobilization of mobile genetic elements called S. aureus pathogenicity islands (SaPIs), such as SaPI1. SaPI1 redirects the assembly pathway of 80α to form capsids that are smaller than those normally made by the phage alone. Both CP and SP of 80α are N-terminally processed by a host-encoded protease, Prp. We have analyzed phage mutants that express pre-cleaved or uncleavable versions of CP or SP, and show that the N-terminal sequence in SP is absolutely required for assembly, but does not need to be cleaved in order to produce viable capsids. Mutants with pre-cleaved or uncleavable CP display normal viability. We have used cryo-EM to solve the structures of mature capsids from an 80α mutant expressing uncleavable CP, and from wildtype SaPI1. Comparisons with structures of 80α and SaPI1 procapsids show that capsid maturation involves major conformational changes in CP, consistent with a release of the CP N-arm by SP. The hexamers reorganize during maturation to accommodate the different environments in the 80α and SaPI1 capsids.


Asunto(s)
Cápside/metabolismo , Fagos de Staphylococcus/fisiología , Staphylococcus aureus/virología , Ensamble de Virus , Cápside/ultraestructura , Microscopía por Crioelectrón , Viabilidad Microbiana , Mutación , Conformación Proteica , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/ultraestructura
16.
Bacteriophage ; 2(2): 70-78, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23050217

RESUMEN

Staphylococcus aureus pathogenicity islands (SaPIs) are mobile genetic elements that encode virulence factors and depend on helper phages for their mobilization. Such mobilization is specific and depends on the ability of a phage protein to inactivate the SaPI repressor Stl. Phage 80α can mobilize several SaPIs, including SaPI1 and SaPIbov1, via its Sri and Dut proteins, respectively. In many cases, the capsids formed in the presence of the SaPI are smaller than those normally produced by the phage. Two SaPI-encoded proteins, CpmA and CpmB, are involved in this size determination process. S. aureus strain Newman contains four prophages, named φNM1 through φNM4. Phages φNM1 and φNM2 are very similar to phage 80α in the structural genes, and encode almost identical Sri proteins, while their Dut proteins are highly divergent. We show that φNM1 and φNM2 are able to mobilize both SaPI1 and SaPIbov1 and yield infectious transducing particles. The majority of the capsids formed in all cases are small, showing that both SaPIs can redirect the capsid size of both φNM1 and φNM2.

17.
Virology ; 432(2): 277-82, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22709958

RESUMEN

SaPIs are molecular pirates that exploit helper bacteriophages for their own high frequency mobilization. One striking feature of helper exploitation by SaPIs is redirection of the phage capsid assembly pathway to produce smaller phage-like particles with T=4 icosahedral symmetry rather than T=7 bacteriophage capsids. Small capsids can accommodate the SaPI genome but not that of the helper phage, leading to interference with helper propagation. Previous studies identified two proteins encoded by the prototype element SaPI1, gp6 and gp7, in SaPI1 procapsids but not in mature SaPI1 particles. Dimers of gp6 form an internal scaffold, aiding fidelity of small capsid assembly. Here we show that both SaPI1 gp6 (CpmB) and gp7 (CpmA) are necessary and sufficient to direct small capsid formation. Surprisingly, failure to form small capsids did not restore wild-type levels of helper phage growth, suggesting an additional role for these SaPI1 proteins in phage interference.


Asunto(s)
Proteínas de la Cápside/metabolismo , Cápside/metabolismo , Islas Genómicas/genética , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/virología , Cápside/ultraestructura , Proteínas de la Cápside/genética , Microscopía por Crioelectrón , Virus Helper/química , Virus Helper/genética , Fagos de Staphylococcus/química , Fagos de Staphylococcus/genética , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Ensamble de Virus
18.
Virology ; 434(2): 242-50, 2012 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22980502

RESUMEN

80α is a temperate, double-stranded DNA bacteriophage of Staphylococcus aureus that can act as a "helper" for the mobilization of S. aureus pathogenicity islands (SaPIs), including SaPI1. When SaPI1 is mobilized by 80α, the SaPI genomes are packaged into capsids that are composed of phage proteins, but that are smaller than those normally formed by the phage. This size determination is dependent on SaPI1 proteins CpmA and CpmB. Here, we show that co-expression of the 80α capsid and scaffolding proteins in S. aureus, but not in E. coli, leads to the formation of procapsid-related structures, suggesting that a host co-factor is required for assembly. The capsid and scaffolding proteins also undergo normal N-terminal processing upon expression in S. aureus, implicating a host protease. We also find that SaPI1 proteins CpmA and CpmB promote the formation of small capsids upon co-expression with 80α capsid and scaffolding proteins in S. aureus.


Asunto(s)
Genética Microbiana/métodos , Biología Molecular/métodos , Fagos de Staphylococcus/fisiología , Staphylococcus aureus/virología , Virología/métodos , Ensamble de Virus , Proteínas Bacterianas/metabolismo , Cápside/metabolismo , Escherichia coli/genética , Expresión Génica , Islas Genómicas , Humanos , Multimerización de Proteína , Fagos de Staphylococcus/genética , Staphylococcus aureus/genética
19.
J Mol Biol ; 405(3): 863-76, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21129380

RESUMEN

Bacteriophages are involved in many aspects of the spread and establishment of virulence factors in Staphylococcus aureus, including the mobilization of genetic elements known as S. aureus pathogenicity islands (SaPIs), which carry genes for superantigen toxins and other virulence factors. SaPIs are packaged into phage-like transducing particles using proteins supplied by the helper phage. We have used cryo-electron microscopy and icosahedral reconstruction to determine the structures of the procapsid and the mature capsid of 80α, a bacteriophage that can mobilize several different SaPIs. The 80α capsid has T=7 icosahedral symmetry with the capsid protein organized into pentameric and hexameric clusters that interact via prominent trimeric densities. The 80α capsid protein was modeled based on the capsid protein fold of bacteriophage HK97 and fitted into the 80α reconstructions. The models show that the trivalent interactions are mediated primarily by a 22-residue ß hairpin structure called the P loop that is not found in HK97. Capsid expansion is associated with a conformational switch in the spine helix that is propagated throughout the subunit, unlike the domain rotation mechanism in phage HK97 or P22.


Asunto(s)
Bacteriófagos/ultraestructura , Proteínas de la Cápside/ultraestructura , Cápside/ultraestructura , Staphylococcus aureus/virología , Secuencia de Aminoácidos , Bacteriófagos/crecimiento & desarrollo , Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Islas Genómicas , Datos de Secuencia Molecular , Conformación Proteica , Staphylococcus aureus/patogenicidad
20.
J Mol Biol ; 412(4): 710-22, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21821042

RESUMEN

Staphylococcus aureus pathogenicity island 1 (SaPI1) is a mobile genetic element that carries genes for several superantigen toxins. SaPI1 is normally stably integrated into the host genome but can become mobilized by "helper" bacteriophage 80α, leading to the packaging of SaPI1 genomes into phage-like transducing particles that are composed of structural proteins supplied by the helper phage but having smaller capsids. We show that the SaPI1-encoded protein gp6 is necessary for efficient formation of small capsids. The NMR structure of gp6 reveals a dimeric protein with a helix-loop-helix motif similar to that of bacteriophage scaffolding proteins. The gp6 dimer matches internal densities that bridge capsid subunits in cryo-electron microscopy reconstructions of SaPI1 procapsids, suggesting that gp6 acts as an internal scaffolding protein in capsid size determination.


Asunto(s)
Proteínas de la Cápside/fisiología , Cápside/fisiología , Tamaño de los Orgánulos/genética , Secuencia de Aminoácidos , Cápside/química , Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Simulación por Computador , Islas Genómicas/genética , Modelos Biológicos , Modelos Moleculares , Organismos Modificados Genéticamente , Pliegue de Proteína , Multimerización de Proteína/genética , Multimerización de Proteína/fisiología , Estructura Cuaternaria de Proteína , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/fisiología , Staphylococcus aureus/ultraestructura , Ensamble de Virus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA