Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Virol ; 97(1): e0133122, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36475765

RESUMEN

Oropouche virus (OROV; genus Orthobunyavirus) is the etiological agent of Oropouche fever, a debilitating febrile illness common in South America. We used recombinant expression of the OROV M polyprotein, which encodes the surface glycoproteins Gn and Gc plus the nonstructural protein NSm, to probe the cellular determinants for OROV assembly and budding. Gn and Gc self-assemble and are secreted independently of NSm. Mature OROV Gn has two predicted transmembrane domains that are crucial for glycoprotein translocation to the Golgi complex and glycoprotein secretion, and unlike related orthobunyaviruses, both transmembrane domains are retained during Gn maturation. Disruption of Golgi function using the drugs brefeldin A and monensin inhibits glycoprotein secretion. Infection studies have previously shown that the cellular endosomal sorting complexes required for transport (ESCRT) machinery is recruited to Golgi membranes during OROV assembly and that ESCRT activity is required for virus secretion. A dominant-negative form of the ESCRT-associated ATPase VPS4 significantly reduces recombinant OROV glycoprotein secretion and blocks virus release from infected cells, and VPS4 partly colocalizes with OROV glycoproteins and membranes costained with Golgi markers. Furthermore, immunoprecipitation and fluorescence microscopy experiments demonstrate that OROV glycoproteins interact with the ESCRT-III component CHMP6, with overexpression of a dominant-negative form of CHMP6 significantly reducing OROV glycoprotein secretion. Taken together, our data highlight differences in M polyprotein processing across orthobunyaviruses, indicate that Golgi and ESCRT function are required for glycoprotein secretion, and identify CHMP6 as an ESCRT-III component that interacts with OROV glycoproteins. IMPORTANCE Oropouche virus causes Oropouche fever, a debilitating illness common in South America that is characterized by high fever, headache, myalgia, and vomiting. The tripartite genome of this zoonotic virus is capable of reassortment, and there have been multiple epidemics of Oropouche fever in South America over the last 50 years, making Oropouche virus infection a significant threat to public health. However, the molecular characteristics of this arbovirus are poorly understood. We developed a recombinant protein expression system to investigate the cellular determinants of OROV glycoprotein maturation and secretion. We show that the proteolytic processing of the M polypeptide, which encodes the surface glycoproteins (Gn and Gc) plus a nonstructural protein (NSm), differs between OROV and its close relative Bunyamwera virus. Furthermore, we demonstrate that OROV M glycoprotein secretion requires the cellular endosomal sorting complexes required for transport (ESCRT) membrane-remodeling machinery and identify that the OROV glycoproteins interact with the ESCRT protein CHMP6.


Asunto(s)
Infecciones por Bunyaviridae , Complejos de Clasificación Endosomal Requeridos para el Transporte , Glicoproteínas de Membrana , Orthobunyavirus , Proteínas Virales , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Orthobunyavirus/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
2.
J Gen Virol ; 102(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33739251

RESUMEN

The enveloped morbilliviruses utilise conserved proteinaceous receptors to enter host cells: SLAMF1 or Nectin-4. Receptor binding is initiated by the viral attachment protein Haemagglutinin (H), with the viral Fusion protein (F) driving membrane fusion. Crystal structures of the prototypic morbillivirus measles virus H with either SLAMF1 or Nectin-4 are available and have served as the basis for improved understanding of this interaction. However, whether these interactions remain conserved throughout the morbillivirus genus requires further characterisation. Using a random mutagenesis approach, based on error-prone PCR, we targeted the putative receptor binding site for SLAMF1 interaction on peste des petits ruminants virus (PPRV) H, identifying mutations that inhibited virus-induced cell-cell fusion. These data, combined with structural modelling of the PPRV H and ovine SLAMF1 interaction, indicate this region is functionally conserved across all morbilliviruses. Error-prone PCR provides a powerful tool for functionally characterising functional domains within viral proteins.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Hemaglutininas Virales/metabolismo , Virus de la Peste de los Pequeños Rumiantes/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Proteínas Virales de Fusión/metabolismo , Animales , Interacciones Microbiota-Huesped , Fusión de Membrana , Ovinos
3.
Science ; 363(6433): 1319-1326, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30765607

RESUMEN

In the 1950s the myxoma virus was released into European rabbit populations in Australia and Europe, decimating populations and resulting in the rapid evolution of resistance. We investigated the genetic basis of resistance by comparing the exomes of rabbits collected before and after the pandemic. We found a strong pattern of parallel evolution, with selection on standing genetic variation favoring the same alleles in Australia, France, and the United Kingdom. Many of these changes occurred in immunity-related genes, supporting a polygenic basis of resistance. We experimentally validated the role of several genes in viral replication and showed that selection acting on an interferon protein has increased the protein's antiviral effect.


Asunto(s)
Adaptación Biológica/genética , Inmunidad Innata/genética , Myxoma virus/inmunología , Mixomatosis Infecciosa/inmunología , Conejos/genética , Conejos/virología , Alelos , Animales , Australia , Evolución Molecular , Francia , Frecuencia de los Genes , Variación Genética , Interferón alfa-2/genética , Interferón alfa-2/inmunología , Mixomatosis Infecciosa/genética , Polimorfismo de Nucleótido Simple , Población , Conejos/inmunología , Reino Unido
4.
Traffic ; 18(1): 44-57, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27813245

RESUMEN

Short peptide motifs in unstructured regions of clathrin-adaptor proteins recruit clathrin to membranes to facilitate post-Golgi membrane transport. Three consensus clathrin-binding peptide sequences have been identified and structural studies show that each binds distinct sites on the clathrin heavy chain N-terminal domain (NTD). A fourth binding site for adaptors on NTD has been functionally identified but not structurally characterised. We have solved high resolution structures of NTD bound to peptide motifs from the cellular clathrin adaptors ß2 adaptin and amphiphysin plus a putative viral clathrin adaptor, hepatitis D virus large antigen (HDAg-L). Surprisingly, with each peptide we observe simultaneous peptide binding at multiple sites on NTD and viral peptides binding to the same sites as cellular peptides. Peptides containing clathrin-box motifs (CBMs) with the consensus sequence LΦxΦ[DE] bind at the 'arrestin box' on NTD, between ß-propeller blades 4 and 5, which had previously been thought to bind a distinct consensus sequence. Further, we structurally define the fourth peptide binding site on NTD, which we term the Royle box. In vitro binding assays show that clathrin is more readily captured by cellular CBMs than by HDAg-L, and site-directed mutagenesis confirms that multiple binding sites on NTD contribute to efficient capture by CBM peptides.


Asunto(s)
Sitios de Unión/fisiología , Cadenas Pesadas de Clatrina/metabolismo , Péptidos/metabolismo , Unión Proteica/fisiología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Antígenos de Hepatitis delta/metabolismo , Humanos , Proteínas del Tejido Nervioso/metabolismo
5.
Viruses ; 7(9): 5084-114, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26393641

RESUMEN

Alphaherpesviruses like herpes simplex virus are large DNA viruses characterized by their ability to establish lifelong latent infection in neurons. As for all herpesviruses, alphaherpesvirus virions contain a protein-rich layer called "tegument" that links the DNA-containing capsid to the glycoprotein-studded membrane envelope. Tegument proteins mediate a diverse range of functions during the virus lifecycle, including modulation of the host-cell environment immediately after entry, transport of virus capsids to the nucleus during infection, and wrapping of cytoplasmic capsids with membranes (secondary envelopment) during virion assembly. Eleven tegument proteins that are conserved across alphaherpesviruses have been implicated in the formation of the tegument layer or in secondary envelopment. Tegument is assembled via a dense network of interactions between tegument proteins, with the redundancy of these interactions making it challenging to determine the precise function of any specific tegument protein. However, recent studies have made great headway in defining the interactions between tegument proteins, conserved across alphaherpesviruses, which facilitate tegument assembly and secondary envelopment. We summarize these recent advances and review what remains to be learned about the molecular interactions required to assemble mature alphaherpesvirus virions following the release of capsids from infected cell nuclei.


Asunto(s)
Alphaherpesvirinae/fisiología , Ensamble de Virus , Modelos Biológicos , Unión Proteica , Proteínas Estructurales Virales/metabolismo
6.
J Biol Chem ; 290(10): 5991-6002, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25605733

RESUMEN

Vaccinia virus (VACV) encodes several proteins that inhibit activation of the proinflammatory transcription factor nuclear factor κB (NF-κB). VACV protein A49 prevents translocation of NF-κB to the nucleus by sequestering cellular ß-TrCP, a protein required for the degradation of the inhibitor of κB. A49 does not share overall sequence similarity with any protein of known structure or function. We solved the crystal structure of A49 from VACV Western Reserve to 1.8 Å resolution and showed, surprisingly, that A49 has the same three-dimensional fold as Bcl-2 family proteins despite lacking identifiable sequence similarity. Whereas Bcl-2 family members characteristically modulate cellular apoptosis, A49 lacks a surface groove suitable for binding BH3 peptides and does not bind proapoptotic Bcl-2 family proteins Bax or Bak. The N-terminal 17 residues of A49 do not adopt a single well ordered conformation, consistent with their proposed role in binding ß-TrCP. Whereas pairs of A49 molecules interact symmetrically via a large hydrophobic surface in crystallo, A49 does not dimerize in solution or in cells, and we propose that this hydrophobic interaction surface may mediate binding to a yet undefined cellular partner. A49 represents the eleventh VACV Bcl-2 family protein and, despite these proteins sharing very low sequence identity, structure-based phylogenetic analysis shows that all poxvirus Bcl-2 proteins are structurally more similar to each other than they are to any cellular or herpesvirus Bcl-2 proteins. This is consistent with duplication and diversification of a single BCL2 family gene acquired by an ancestral poxvirus.


Asunto(s)
Inmunidad Innata/genética , Filogenia , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Virales/química , Apoptosis/genética , Cristalografía por Rayos X , Células HEK293 , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Conformación Proteica , Pliegue de Proteína , Vaccinia/genética , Vaccinia/virología , Virus Vaccinia/química , Virus Vaccinia/genética , Virus Vaccinia/patogenicidad , Proteínas Virales/genética
7.
Science ; 345(6195): 459-63, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-25061211

RESUMEN

Clathrin-mediated endocytosis (CME) is vital for the internalization of most cell-surface proteins. In CME, plasma membrane-binding clathrin adaptors recruit and polymerize clathrin to form clathrin-coated pits into which cargo is sorted. Assembly polypeptide 2 (AP2) is the most abundant adaptor and is pivotal to CME. Here, we determined a structure of AP2 that includes the clathrin-binding ß2 hinge and developed an AP2-dependent budding assay. Our findings suggest that an autoinhibitory mechanism prevents clathrin recruitment by cytosolic AP2. A large-scale conformational change driven by the plasma membrane phosphoinositide phosphatidylinositol 4,5-bisphosphate and cargo relieves this autoinhibition, triggering clathrin recruitment and hence clathrin-coated bud formation. This molecular switching mechanism can couple AP2's membrane recruitment to its key functions of cargo and clathrin binding.


Asunto(s)
Complejo 2 de Proteína Adaptadora/química , Subunidades beta de Complejo de Proteína Adaptadora/química , Membrana Celular/química , Clatrina/química , Polimerizacion , Endocitosis , Humanos , Fosfatidilinositol 4,5-Difosfato/química
8.
Proc Natl Acad Sci U S A ; 110(47): E4482-91, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24194549

RESUMEN

Mutations in either syntaxin 11 (Stx11) or Munc18-2 abolish cytotoxic T lymphocytes (CTL) and natural killer cell (NK) cytotoxicity, and give rise to familial hemophagocytic lymphohistiocytosis (FHL4 or FHL5, respectively). Although Munc18-2 is known to interact with Stx11, little is known about the molecular mechanisms governing the specificity of this interaction or how in vitro IL-2 activation leads to compensation of CTL and NK cytotoxicity. To understand how mutations in Munc18-2 give rise to disease, we have solved the structure of human Munc18-2 at 2.6 Å resolution and mapped 18 point mutations. The four surface mutations identified (R39P, L130S, E132A, P334L) map exclusively to the predicted syntaxin and soluble N-ethylmaleimide-sensitive factor accessory protein receptor binding sites of Munc18-2. We find that Munc18-2 binds the N-terminal peptide of Stx11 with a ~20-fold higher affinity than Stx3, suggesting a potential role in selective binding. Upon IL-2 activation, levels of Stx3 are increased, favoring Munc18-2 binding when Stx11 is absent. Similarly, Munc18-1, expressed in IL-2-activated CTL, is capable of binding Stx11. These findings provide potential explanations for restoration of Munc18-Stx function and cytotoxicity in IL-2-activated cells.


Asunto(s)
Evolución Molecular , Células Asesinas Naturales/inmunología , Linfohistiocitosis Hemofagocítica/genética , Modelos Moleculares , Proteínas Munc18/química , Proteínas Qa-SNARE/metabolismo , Linfocitos T Citotóxicos/inmunología , Animales , Western Blotting , Cristalización , Células HEK293 , Humanos , Inmunohistoquímica , Células Asesinas Naturales/metabolismo , Proteínas Munc18/genética , Proteínas Munc18/metabolismo , Mutación Puntual/genética , Unión Proteica , Células Sf9 , Spodoptera , Linfocitos T Citotóxicos/metabolismo
9.
J Virol ; 87(6): 3229-36, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23283942

RESUMEN

Infectious pancreatic necrosis virus (IPNV), a member of the family Birnaviridae, infects young salmon, with a severe impact on the commercial sea farming industry. Of the five mature proteins encoded by the IPNV genome, the multifunctional VP3 has an essential role in morphogenesis; interacting with the capsid protein VP2, the viral double-stranded RNA (dsRNA) genome and the RNA-dependent RNA polymerase VP1. Here we investigate one of these VP3 functions and present the crystal structure of the C-terminal 12 residues of VP3 bound to the VP1 polymerase. This interaction, visualized for the first time, reveals the precise molecular determinants used by VP3 to bind the polymerase. Competition binding studies confirm that this region of VP3 is necessary and sufficient for VP1 binding, while biochemical experiments show that VP3 attachment has no effect on polymerase activity. These results indicate how VP3 recruits the polymerase into birnavirus capsids during morphogenesis.


Asunto(s)
Virus de la Necrosis Pancreática Infecciosa/química , ARN Polimerasa Dependiente del ARN/química , Proteínas Estructurales Virales/química , Cristalografía por Rayos X , Unión Proteica , Conformación Proteica , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Estructurales Virales/metabolismo
10.
PLoS Pathog ; 7(12): e1002430, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22194685

RESUMEN

Vaccinia virus (VACV) protein N1 is an intracellular virulence factor and belongs to a family of VACV B-cell lymphoma (Bcl)-2-like proteins whose members inhibit apoptosis or activation of pro-inflammatory transcription factors, such as interferon (IFN) regulatory factor-3 (IRF-3) and nuclear factor-κB (NF-κB). Unusually, N1 inhibits both apoptosis and NF-κB activation. To understand how N1 exerts these different functions, we have mutated residues in the Bcl-2-like surface groove and at the interface used to form N1 homodimers. Mutagenesis of the surface groove abolished only the N1 anti-apoptotic activity and protein crystallography showed these mutants differed from wild-type N1 only at the site of mutation. Conversely, mutagenesis of the dimer interface converted N1 to a monomer and affected only inhibition of NF-κB activation. Collectively, these data show that N1 inhibits pro-inflammatory and pro-apoptotic signalling using independent surfaces of the protein. To determine the relative contribution of each activity to virus virulence, mutant N1 alleles were introduced into a VACV strain lacking N1 and the virulence of these viruses was analysed after intradermal and intranasal inoculation in mice. In both models, VACV containing a mutant N1 unable to inhibit apoptosis had similar virulence to wild-type virus, whereas VACV containing a mutant N1 impaired for NF-κB inhibition induced an attenuated infection similar to that of the N1-deleted virus. This indicates that anti-apoptotic activity of N1 does not drive virulence in these in vivo models, and highlights the importance of pro-inflammatory signalling in the immune response against viral infections.


Asunto(s)
Apoptosis/fisiología , FN-kappa B/metabolismo , Virus Vaccinia/patogenicidad , Proteínas Virales/química , Proteínas Virales/metabolismo , Animales , Línea Celular , Humanos , Ratones , Mutación/genética , Unión Proteica , Estructura Terciaria de Proteína , Virus Vaccinia/genética , Virus Vaccinia/metabolismo , Proteínas Virales/genética , Virulencia
11.
PLoS Pathog ; 7(6): e1002085, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21731487

RESUMEN

The RNA-dependent RNA polymerase VP1 of infectious pancreatic necrosis virus (IPNV) is a single polypeptide responsible for both viral RNA transcription and genome replication. Sequence analysis identifies IPNV VP1 as having an unusual active site topology. We have purified, crystallized and solved the structure of IPNV VP1 to 2.3 Å resolution in its apo form and at 2.2 Å resolution bound to the catalytically-activating metal magnesium. We find that recombinantly-expressed VP1 is highly active for RNA transcription and replication, yielding both free and polymerase-attached RNA products. IPNV VP1 also possesses terminal (deoxy)nucleotide transferase, RNA-dependent DNA polymerase (reverse transcriptase) and template-independent self-guanylylation activity. The N-terminus of VP1 interacts with the active-site cleft and we show that the N-terminal serine residue is required for formation of covalent RNA:polymerase complexes, providing a mechanism for the genesis of viral genome:polymerase complexes observed in vivo.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/química , Genoma Viral , Virus de la Necrosis Pancreática Infecciosa/enzimología , Dominio Catalítico , Cristalografía por Rayos X , ARN Polimerasas Dirigidas por ADN/metabolismo , Magnesio , Unión Proteica , Conformación Proteica , ARN Viral/biosíntesis , Transcripción Genética
12.
Structure ; 19(7): 1011-20, 2011 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-21742267

RESUMEN

The morphogenesis of poxviruses such as vaccinia virus (VACV) sees the virion shape mature from spherical to brick-shaped. Trimeric capsomers of the VACV D13 protein form a transitory, stabilizing lattice on the surface of the initial spherical immature virus particle. The crystal structure of D13 reveals that this major scaffolding protein comprises a double ß barrel "jelly-roll" subunit arranged as pseudo-hexagonal trimers. These structural features are characteristic of the major capsid proteins of a lineage of large icosahedral double-stranded DNA viruses including human adenovirus and the bacteriophages PRD1 and PM2. Structure-based phylogenetic analysis confirms that VACV belongs to this lineage, suggesting that (analogously to higher organism embryogenesis) early poxvirus morphogenesis reflects their evolution from a lineage of viruses sharing a common icosahedral ancestor.


Asunto(s)
Proteínas de la Cápside/química , Cápside/química , Proteínas Recombinantes de Fusión/química , Virus Vaccinia/química , Secuencia de Aminoácidos , Bacteriófago PRD1/química , Evolución Biológica , Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Clonación Molecular , Corticoviridae/química , Cristalización , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Plásmidos , Conformación Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Vaccinia/virología , Virus Vaccinia/genética , Virus Vaccinia/metabolismo
13.
J Struct Biol ; 175(2): 127-34, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21419849

RESUMEN

Viruses are obligate intracellular parasites and are some of the most rapidly evolving and diverse pathogens encountered by the host immune system. Large complicated viruses, such as poxviruses, have evolved a plethora of proteins to disrupt host immune signalling in their battle against immune surveillance. Recent X-ray crystallographic analysis of these viral immunomodulators has helped form an emerging picture of the molecular details of virus-host interactions. In this review we consider some of these immune evasion strategies as they apply to poxviruses, from a structural perspective, with specific examples from the European SPINE2-Complexes initiative. Structures of poxvirus immunomodulators reveal the capacity of viruses to mimic and compete against the host immune system, using a diverse range of structural folds that are unique or acquired from their hosts with both enhanced and unexpectedly divergent functions.


Asunto(s)
Evolución Biológica , Evasión Inmune , Virus Vaccinia/fisiología , Proteínas Virales/química , Secuencia de Aminoácidos , Animales , Quimiocinas/antagonistas & inhibidores , Quimiocinas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Factores Inmunológicos/química , Factores Inmunológicos/metabolismo , Datos de Secuencia Molecular , Filogenia , Poxviridae/genética , Poxviridae/inmunología , Poxviridae/fisiología , Conformación Proteica , Transducción de Señal , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Proteínas Virales/metabolismo
14.
Nucleic Acids Res ; 39(9): 3652-66, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21245041

RESUMEN

Ribonuclease H2 is the major nuclear enzyme degrading cellular RNA/DNA hybrids in eukaryotes and the sole nuclease known to be able to hydrolyze ribonucleotides misincorporated during genomic replication. Mutation in RNASEH2 causes Aicardi-Goutières syndrome, an auto-inflammatory disorder that may arise from nucleic acid byproducts generated during DNA replication. Here, we report the crystal structures of Archaeoglobus fulgidus RNase HII in complex with PCNA, and human PCNA bound to a C-terminal peptide of RNASEH2B. In the archaeal structure, three binding modes are observed as the enzyme rotates about a flexible hinge while anchored to PCNA by its PIP-box motif. PCNA binding promotes RNase HII activity in a hinge-dependent manner. It enhances both cleavage of ribonucleotides misincorporated in DNA duplexes, and the comprehensive hydrolysis of RNA primers formed during Okazaki fragment maturation. In addition, PCNA imposes strand specificity on enzyme function, and by localizing RNase H2 and not RNase H1 to nuclear replication foci in vivo it ensures that RNase H2 is the dominant RNase H activity during nuclear replication. Our findings provide insights into how type 2 RNase H activity is directed during genome replication and repair, and suggest a mechanism by which RNase H2 may suppress generation of immunostimulatory nucleic acids.


Asunto(s)
Replicación del ADN , Antígeno Nuclear de Célula en Proliferación/química , Ribonucleasa H/química , Archaeoglobus fulgidus/enzimología , Cristalografía , Enzimas Reparadoras del ADN/química , Enzimas Reparadoras del ADN/metabolismo , Humanos , Modelos Moleculares , Péptidos/química , Ribonucleasa H/metabolismo
15.
J Biol Chem ; 284(39): 26613-9, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19628875

RESUMEN

Signal-regulatory protein alpha (SIRPalpha) is a myeloid membrane receptor that interacts with the membrane protein CD47, a marker of self. We have solved the structure of the complete extracellular portion of SIRPalpha, comprising three immunoglobulin superfamily domains, by x-ray crystallography to 2.5 A resolution. These data, together with previous data on the N-terminal domain and its ligand CD47 (possessing a single immunoglobulin superfamily domain), show that the CD47-SIRPalpha interaction will span a distance of around 14 nm between interacting cells, comparable with that of an immunological synapse. The N-terminal (V-set) domain mediates binding to CD47, and the two others are found to be constant (C1-set) domains. C1-set domains are restricted to proteins involved in vertebrate antigen recognition: T cell antigen receptors, immunoglobulins, major histocompatibility complex antigens, tapasin, and beta2-microglobulin. The domains of SIRPalpha (domains 2 and 3) are structurally more similar to C1-set domains than any cell surface protein not involved in antigen recognition. This strengthens the suggestion from sequence analysis that SIRP is evolutionarily closely related to antigen recognition proteins.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Antígeno CD47/metabolismo , Receptores Inmunológicos/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos de Diferenciación/química , Antígenos de Diferenciación/genética , Sitios de Unión/genética , Antígeno CD47/química , Células CHO , Análisis por Conglomerados , Cricetinae , Cricetulus , Cristalografía por Rayos X , Evolución Molecular , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Homología de Secuencia de Aminoácido
16.
PLoS Pathog ; 4(8): e1000128, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18704168

RESUMEN

Vaccinia virus (VACV), the prototype poxvirus, encodes numerous proteins that modulate the host response to infection. Two such proteins, B14 and A52, act inside infected cells to inhibit activation of NF-kappaB, thereby blocking the production of pro-inflammatory cytokines. We have solved the crystal structures of A52 and B14 at 1.9 A and 2.7 A resolution, respectively. Strikingly, both these proteins adopt a Bcl-2-like fold despite sharing no significant sequence similarity with other viral or cellular Bcl-2-like proteins. Unlike cellular and viral Bcl-2-like proteins described previously, A52 and B14 lack a surface groove for binding BH3 peptides from pro-apoptotic Bcl-2-like proteins and they do not modulate apoptosis. Structure-based phylogenetic analysis of 32 cellular and viral Bcl-2-like protein structures reveals that A52 and B14 are more closely related to each other and to VACV N1 and myxoma virus M11 than they are to other viral or cellular Bcl-2-like proteins. This suggests that a progenitor poxvirus acquired a gene encoding a Bcl-2-like protein and, over the course of evolution, gene duplication events have allowed the virus to exploit this Bcl-2 scaffold for interfering with distinct host signalling pathways.


Asunto(s)
Apoptosis , Evolución Molecular , FN-kappa B/química , Proteínas Proto-Oncogénicas c-bcl-2/química , Virus Vaccinia/química , Proteínas Virales/química , Línea Celular , Cristalografía por Rayos X , Humanos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Pliegue de Proteína , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Vaccinia/metabolismo , Virus Vaccinia/metabolismo , Proteínas Virales/metabolismo
17.
Mol Cell ; 31(2): 266-77, 2008 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-18657508

RESUMEN

CD47 is a widely distributed cell-surface protein that acts a marker of self through interactions of myeloid and neural cells. We describe the high-resolution X-ray crystallographic structures of the immunoglobulin superfamily domain of CD47 alone and in complex with the N-terminal ligand-binding domain of signal regulatory protein alpha (SIRPalpha). The unusual and convoluted interacting face of CD47, comprising the N terminus and loops at the end of the domain, intercalates with the corresponding regions in SIRPalpha. We have also determined structures of the N-terminal domains of SIRPbeta, SIRPbeta(2), and SIRPgamma; proteins that are closely related to SIRPalpha but bind CD47 with negligible or reduced affinity. These results explain the specificity of CD47 for the SIRP family of paired receptors in atomic detail. Analysis of SIRPalpha polymorphisms suggests that these, as well as the activating SIRPs, may have evolved to counteract pathogen binding to the inhibitory SIRPalpha receptor.


Asunto(s)
Antígeno CD47/química , Receptores Inmunológicos/química , Receptores Inmunológicos/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Comunicación Celular , Cricetinae , Cricetulus , Cristalografía por Rayos X , Macrófagos/citología , Modelos Moleculares , Datos de Secuencia Molecular , Polimorfismo Genético , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
18.
Arch Biochem Biophys ; 469(2): 200-8, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17983589

RESUMEN

Aminopeptidase P (APPro) is a manganese-containing enzyme that catalyses the hydrolysis of the N-terminal residue of a polypeptide if the second residue is proline. Structures of APPro mutants with reduced or negligible activity have been determined in complex with the tripeptide substrate ValProLeu. In the complex of Glu383Ala APPro with ValProLeu one of the two metal sites is only partly occupied, indicating an essential role for Glu383 in metal binding in the presence of substrate. His361Ala APPro clearly possesses residual activity as the ValProLeu substrate has been cleaved in the crystals; difference electron density consistent with bound ProLeu dipeptide and a disordered Val amino acid is present at the active site. Contrary to previous suggestions, the His243Ala mutant is capable of binding substrate. The structure of the His243Ala APPro complex with ValProLeu shows that the peptide interacts with one of the active-site metal atoms via its terminal amino group. The implications of these complexes for the roles of the respective residues in APPro catalysis are discussed.


Asunto(s)
Aminopeptidasas/genética , Escherichia coli/enzimología , Escherichia coli/genética , Mutación , Sitios de Unión , Bioquímica/métodos , Cristalografía por Rayos X/métodos , Leucina/química , Modelos Químicos , Conformación Molecular , Péptidos/química , Prolina/química , Especificidad por Sustrato , Valina/química
19.
J Mol Biol ; 372(3): 660-71, 2007 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-17681535

RESUMEN

Vaccinia virus (VACV), the smallpox vaccine, encodes many proteins that subvert the host immune response. One of these, cytokine response modifier E (CrmE), is secreted by infected cells and protects these cells from apoptotic challenge by tumour necrosis factor alpha (TNFalpha). We have expressed recombinant CrmE from VACV strain Lister in Escherichia coli, shown that the purified protein is monomeric in solution and competent to bind TNFalpha, and solved the structure to 2.0 A resolution. This is the first structure of a virus-encoded tumour necrosis factor receptor (TNFR). CrmE shares significant sequence similarity with mammalian type 2 TNF receptors (TNFSFR1B, p75; TNFR type 2). The structure confirms that CrmE adopts the canonical TNFR fold but only one of the two "ligand-binding" loops of TNFRSF1A is conserved in CrmE, suggesting a mechanism for the higher affinity of poxvirus TNFRs for TNFalpha over lymphotoxin-alpha. The roles of dimerisation and pre-ligand-assembly domains (PLADs) in poxvirus and mammalian TNFR activity are discussed.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/química , Virus Vaccinia/química , Proteínas Virales/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Receptores del Factor de Necrosis Tumoral/aislamiento & purificación , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/química , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Factor de Necrosis Tumoral alfa/aislamiento & purificación , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Virales/aislamiento & purificación , Proteínas Virales/metabolismo
20.
Biochemistry ; 45(3): 964-75, 2006 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-16411772

RESUMEN

Aminopeptidase P (APPro) is a manganese-dependent enzyme that cleaves the N-terminal amino acid from polypeptides where the second residue is proline. APPro shares a similar fold, substrate specificity, and catalytic mechanism with methionine aminopeptidase and prolidase. To investigate the roles of conserved residues at the active site, seven mutant forms of APPro were characterized kinetically and structurally. Mutation of individual metal ligands selectively abolished binding of either or both Mn(II) atoms at the active site, and none of these metal-ligand mutants had detectable catalytic activity. Mutation of the conserved active site residues His243 and His361 revealed that both are required for catalysis. We propose that His243 stabilizes substrate binding through an interaction with the carbonyl oxygen of the requisite proline residue of a substrate and that His361 stabilizes substrate binding and the gem-diol catalytic intermediate. Sequence, structural, and kinetic analyses reveal that His350, conserved in APPro and prolidase but not in methionine aminopeptidase, forms part of a hydrophobic binding pocket that gives APPro its proline specificity. Further, peptides in which the required proline residue is replaced by N-methylalanine or alanine are cleaved by APPro, but they are extremely poor substrates due to a loss of interactions between the prolidyl ring of the substrate and the hydrophobic proline-binding pocket.


Asunto(s)
Aminopeptidasas/química , Aminopeptidasas/metabolismo , Escherichia coli/enzimología , Mutación/genética , Aminopeptidasas/genética , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Escherichia coli/genética , Cinética , Modelos Moleculares , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA