Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Invertebr Pathol ; 186: 107570, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33775676

RESUMEN

Organisms have evolved mechanisms in which cellular membranes can both be targeted and punctured thereby killing the targeted cell. One such mechanism involves the deployment of pore forming proteins (PFPs) which function by oligomerizing on cell membranes and inserting a physical pore spanning the membrane. This pore can lead to cell death by either causing osmotic flux or allowing the delivery of a secondary toxin. Pore forming proteins can be broadly classified into different families depending on the structure of the final pore; either α-PFPs using channels made from α -helices or ß-PFPs using channels made from ß-barrels. There are many different ß-PFPs and an emerging superfamily is the aerolysin-ETX/MTX-2 superfamily. A comparison between the members of this superfamily reveals the pore forming domain is a common module yet the receptor binding region is highly variable. These structural and architectural variations lead to differences in the target recognition and determine the site of activity. Closer investigation of the topology of the family also suggests that the Toxin_10 family of PFPs could be considered as part of the aerolysin-ETX/MTX-2 superfamily. Comparatively, far less is known about how Toxin_10 proteins assemble into the final pore structure than aerolysin-ETX/MTX-2 proteins. This review aims to collate the pore forming protein members and bridge the structural similarities between the aerolysin-ETX/MTX-2 superfamily and the insecticidal Toxin_10 subfamily.


Asunto(s)
Bacterias/química , Toxinas Bacterianas/química , Proteínas Citotóxicas Formadoras de Poros/química
2.
Anal Chem ; 92(7): 5168-5177, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32155328

RESUMEN

Unravelling the three-dimensional structures and compositions of biological macromolecules sheds light on their functions and also contributes to the design of future biochemical compounds and processes. Atom probe tomography (APT) is demonstrated in this research as a new and effective approach to explore the structure and chemical composition of a single protein in the hydrated state. By introducing graphene encapsulation, proteins in solution can be immobilized on a metal specimen tip, with an end radius in the range of 50 nm to allow field ionization and evaporation. Using a ferritin particle as an example, analysis of the mass spectrum and reconstructed 3D chemical maps at near-atomic resolution acquired from APT reveals the core consisting of iron and iron oxides, the peptide shell containing amino acids, and the interior interface between the iron core and the peptide shell. The quantitative distribution and proportion of iron isotopes from a single ferritin core have been determined for the first time, as well as identification of the possible sites of amino acids inside the protein shell. The complete experimental protocol is straightforward and lays a foundation for future exploration of various macromolecules in a controlled environment.


Asunto(s)
Ferritinas/análisis , Tomografía Computarizada por Rayos X , Animales , Grafito/química , Caballos , Bazo/química
3.
Mol Cell Proteomics ; 17(12): 2324-2334, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30097534

RESUMEN

Esophageal adenocarcinoma (EAC) is thought to develop from asymptomatic Barrett's esophagus (BE) with a low annual rate of conversion. Current endoscopy surveillance of BE patients is probably not cost-effective. Previously, we discovered serum glycoprotein biomarker candidates which could discriminate BE patients from EAC. Here, we aimed to validate candidate serum glycoprotein biomarkers in independent cohorts, and to develop a biomarker candidate panel for BE surveillance. Serum glycoprotein biomarker candidates were measured in 301 serum samples collected from Australia (4 states) and the United States (1 clinic) using previously established lectin magnetic bead array (LeMBA) coupled multiple reaction monitoring mass spectrometry (MRM-MS) tier 3 assay. The area under receiver operating characteristic curve (AUROC) was calculated as a measure of discrimination, and multivariate recursive partitioning was used to formulate a multi-marker panel for BE surveillance. Complement C9 (C9), gelsolin (GSN), serum paraoxonase/arylesterase 1 (PON1) and serum paraoxonase/lactonase 3 (PON3) were validated as diagnostic glycoprotein biomarkers in lectin pull-down samples for EAC across both cohorts. A panel of 10 serum glycoprotein biomarker candidates discriminated BE patients not requiring intervention (BE± low grade dysplasia) from those requiring intervention (BE with high grade dysplasia (BE-HGD) or EAC) with an AUROC value of 0.93. Tissue expression of C9 was found to be induced in BE, dysplastic BE and EAC. In longitudinal samples from subjects that have progressed toward EAC, levels of serum C9 were significantly (p < 0.05) increased with disease progression in EPHA (erythroagglutinin from Phaseolus vulgaris) and NPL (Narcissus pseudonarcissus lectin) pull-down samples. The results confirm alteration of complement pathway glycoproteins during BE-EAC pathogenesis. Further prospective clinical validation of the confirmed biomarker candidates in a large cohort is warranted, prior to development of a first-line BE surveillance blood test.


Asunto(s)
Adenocarcinoma/sangre , Arildialquilfosfatasa/sangre , Esófago de Barrett/sangre , Complemento C9/análisis , Neoplasias Esofágicas/sangre , Gelsolina/sangre , Adenocarcinoma/diagnóstico , Adenocarcinoma/etiología , Adenocarcinoma/patología , Anciano , Área Bajo la Curva , Australia , Esófago de Barrett/complicaciones , Esófago de Barrett/diagnóstico , Esófago de Barrett/patología , Biomarcadores/sangre , Biopsia , Estudios de Cohortes , Diagnóstico Diferencial , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/etiología , Neoplasias Esofágicas/patología , Femenino , Humanos , Masculino , Espectrometría de Masas/métodos , Persona de Mediana Edad , Análisis Multivariante , Vigilancia en Salud Pública , Estados Unidos
4.
Immunity ; 30(6): 777-88, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19464197

RESUMEN

Ligation of the alphabeta T cell receptor (TCR) by a specific peptide-loaded major histocompatibility complex (pMHC) molecule initiates T cell signaling via the CD3 complex. However, the initial events that link antigen recognition to T cell signal transduction remain unclear. Here we show, via fluorescence-based experiments and structural analyses, that MHC-restricted antigen recognition by the alphabeta TCR results in a specific conformational change confined to the A-B loop within the alpha chain of the constant domain (Calpha). The apparent affinity constant of this A-B loop movement mirrored that of alphabeta TCR-pMHC ligation and was observed in two alphabeta TCRs with distinct pMHC specificities. The Ag-induced A-B loop conformational change could be inhibited by fixing the juxtapositioning of the constant domains and was shown to be reversible upon pMHC disassociation. Notably, the loop movement within the Calpha domain, although specific for an agonist pMHC ligand, was not observed with a pMHC antagonist. Moreover, mutagenesis of residues within the A-B loop impaired T cell signaling in an in vitro system of antigen-specific TCR stimulation. Collectively, our findings provide a basis for the earliest molecular events that underlie Ag-induced T cell triggering.


Asunto(s)
Antígenos/química , Receptores de Antígenos de Linfocitos T alfa-beta/química , Linfocitos T/inmunología , Animales , Antígenos/inmunología , Humanos , Complejo Mayor de Histocompatibilidad/inmunología , Mutación/genética , Péptidos/química , Péptidos/inmunología , Unión Proteica/inmunología , Estructura Terciaria de Proteína , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología
5.
PLoS Biol ; 13(2): e1002049, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25654333

RESUMEN

Membrane attack complex/perforin-like (MACPF) proteins comprise the largest superfamily of pore-forming proteins, playing crucial roles in immunity and pathogenesis. Soluble monomers assemble into large transmembrane pores via conformational transitions that remain to be structurally and mechanistically characterised. Here we present an 11 Å resolution cryo-electron microscopy (cryo-EM) structure of the two-part, fungal toxin Pleurotolysin (Ply), together with crystal structures of both components (the lipid binding PlyA protein and the pore-forming MACPF component PlyB). These data reveal a 13-fold pore 80 Å in diameter and 100 Å in height, with each subunit comprised of a PlyB molecule atop a membrane bound dimer of PlyA. The resolution of the EM map, together with biophysical and computational experiments, allowed confident assignment of subdomains in a MACPF pore assembly. The major conformational changes in PlyB are a ∼70° opening of the bent and distorted central ß-sheet of the MACPF domain, accompanied by extrusion and refolding of two α-helical regions into transmembrane ß-hairpins (TMH1 and TMH2). We determined the structures of three different disulphide bond-trapped prepore intermediates. Analysis of these data by molecular modelling and flexible fitting allows us to generate a potential trajectory of ß-sheet unbending. The results suggest that MACPF conformational change is triggered through disruption of the interface between a conserved helix-turn-helix motif and the top of TMH2. Following their release we propose that the transmembrane regions assemble into ß-hairpins via top down zippering of backbone hydrogen bonds to form the membrane-inserted ß-barrel. The intermediate structures of the MACPF domain during refolding into the ß-barrel pore establish a structural paradigm for the transition from soluble monomer to pore, which may be conserved across the whole superfamily. The TMH2 region is critical for the release of both TMH clusters, suggesting why this region is targeted by endogenous inhibitors of MACPF function.


Asunto(s)
Membrana Celular/química , Complejo de Ataque a Membrana del Sistema Complemento/química , Proteínas Fúngicas/química , Proteínas Hemolisinas/química , Pleurotus/química , Proteínas Recombinantes de Fusión/química , Animales , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Eritrocitos/química , Eritrocitos/citología , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Expresión Génica , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Modelos Moleculares , Unión Proteica , Pliegue de Proteína , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Ovinos
6.
Proc Natl Acad Sci U S A ; 112(50): 15360-5, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26627714

RESUMEN

The lethal factor in stonefish venom is stonustoxin (SNTX), a heterodimeric cytolytic protein that induces cardiovascular collapse in humans and native predators. Here, using X-ray crystallography, we make the unexpected finding that SNTX is a pore-forming member of an ancient branch of the Membrane Attack Complex-Perforin/Cholesterol-Dependent Cytolysin (MACPF/CDC) superfamily. SNTX comprises two homologous subunits (α and ß), each of which comprises an N-terminal pore-forming MACPF/CDC domain, a central focal adhesion-targeting domain, a thioredoxin domain, and a C-terminal tripartite motif family-like PRY SPla and the RYanodine Receptor immune recognition domain. Crucially, the structure reveals that the two MACPF domains are in complex with one another and arranged into a stable early prepore-like assembly. These data provide long sought after near-atomic resolution insights into how MACPF/CDC proteins assemble into prepores on the surface of membranes. Furthermore, our analyses reveal that SNTX-like MACPF/CDCs are distributed throughout eukaryotic life and play a broader, possibly immune-related function outside venom.


Asunto(s)
Venenos de los Peces/química , Perforina/química , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Colesterol/química , Complejo de Ataque a Membrana del Sistema Complemento/química , Cristalografía por Rayos X , Microscopía Electrónica de Transmisión , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Solubilidad , Homología Estructural de Proteína
7.
Biochim Biophys Acta ; 1858(3): 475-86, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26607011

RESUMEN

Pore Forming Toxins (PFTs) represent a key mechanism for permitting the passage of proteins and small molecules across the lipid membrane. These proteins are typically produced as soluble monomers that self-assemble into ring-like oligomeric structures on the membrane surface. Following such assembly PFTs undergo a remarkable conformational change to insert into the lipid membrane. While many different protein families have independently evolved such ability, members of the Membrane Attack Complex PerForin/Cholesterol Dependent Cytolysin (MACPF/CDC) superfamily form distinctive giant ß-barrel pores comprised of up to 50 monomers and up to 300Å in diameter. In this review we focus on recent advances in understanding the structure of these giant MACPF/CDC pores as well as the underlying molecular mechanisms leading to their formation. Commonalities and evolved variations of the pore forming mechanism across the superfamily are discussed. This article is part of a Special Issue entitled: Pore-Forming Toxins edited by Mauro Dalla Serra and Franco Gambale.


Asunto(s)
Membrana Celular/metabolismo , Evolución Molecular , Perforina/clasificación , Perforina/metabolismo , Animales , Membrana Celular/química , Humanos , Perforina/química , Estructura Secundaria de Proteína , Relación Estructura-Actividad
8.
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
9.
J Biol Chem ; 289(13): 9172-81, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24558045

RESUMEN

Cytotoxic lymphocytes eliminate virally infected or neoplastic cells through the action of cytotoxic proteases (granzymes). The pore-forming protein perforin is essential for delivery of granzymes into the cytoplasm of target cells; however the mechanism of this delivery is incompletely understood. Perforin contains a membrane attack complex/perforin (MACPF) domain and oligomerizes to form an aqueous pore in the plasma membrane; therefore the simplest (and best supported) model suggests that granzymes passively diffuse through the perforin pore into the cytoplasm of the target cell. Here we demonstrate that perforin preferentially delivers cationic molecules while anionic and neutral cargoes are delivered inefficiently. Furthermore, another distantly related pore-forming MACPF protein, pleurotolysin (from the oyster mushroom), also favors the delivery of cationic molecules, and efficiently delivers human granzyme B. We propose that this facilitated diffusion is due to conserved features of oligomerized MACPF proteins, which may include an anionic lumen.


Asunto(s)
Perforina/química , Perforina/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Transporte Biológico , Cationes/metabolismo , Línea Celular , Difusión , Granzimas/metabolismo , Proteínas Hemolisinas/metabolismo , Heparina/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Porosidad , Estructura Terciaria de Proteína
10.
J Biol Chem ; 289(40): 27979-91, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25135637

RESUMEN

Cysteine-containing peptides represent an important class of T cell epitopes, yet their prevalence remains underestimated. We have established and interrogated a database of around 70,000 naturally processed MHC-bound peptides and demonstrate that cysteine-containing peptides are presented on the surface of cells in an MHC allomorph-dependent manner and comprise on average 5-10% of the immunopeptidome. A significant proportion of these peptides are oxidatively modified, most commonly through covalent linkage with the antioxidant glutathione. Unlike some of the previously reported cysteine-based modifications, this represents a true physiological alteration of cysteine residues. Furthermore, our results suggest that alterations in the cellular redox state induced by viral infection are communicated to the immune system through the presentation of S-glutathionylated viral peptides, resulting in altered T cell recognition. Our data provide a structural basis for how the glutathione modification alters recognition by virus-specific T cells. Collectively, these results suggest that oxidative stress represents a mechanism for modulating the virus-specific T cell response.


Asunto(s)
Presentación de Antígeno , Infecciones por Coronavirus/veterinaria , Epítopos de Linfocito T/metabolismo , Virus de la Hepatitis Murina/inmunología , Enfermedades de los Roedores/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/virología , Linfocitos T CD8-positivos/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Cisteína/metabolismo , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Glutatión/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/genética , Oxidación-Reducción , Enfermedades de los Roedores/inmunología , Enfermedades de los Roedores/virología
11.
Nat Chem Biol ; 9(10): 605-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24045807

RESUMEN

Cryo-EM, crystallography, biochemical experiments and computational approaches have been used to study different intermediate states of the Aeromonas hydrophila toxin aerolysin en route to pore formation. These results reveal that an unexpected and marked rotation of the core aerolysin machinery is required to unleash the membrane-spanning regions.


Asunto(s)
Aeromonas/química , Toxinas Bacterianas/química , Canales Iónicos/química , Precursores de Proteínas/química
12.
PLoS Comput Biol ; 10(8): e1003791, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25144725

RESUMEN

Cholesterol Dependent Cytolysins (CDCs) are important bacterial virulence factors that form large (200-300 Å) membrane embedded pores in target cells. Currently, insights from X-ray crystallography, biophysical and single particle cryo-Electron Microscopy (cryo-EM) experiments suggest that soluble monomers first interact with the membrane surface via a C-terminal Immunoglobulin-like domain (Ig; Domain 4). Membrane bound oligomers then assemble into a prepore oligomeric form, following which the prepore assembly collapses towards the membrane surface, with concomitant release and insertion of the membrane spanning subunits. During this rearrangement it is proposed that Domain 2, a region comprising three ß-strands that links the pore forming region (Domains 1 and 3) and the Ig domain, must undergo a significant yet currently undetermined, conformational change. Here we address this problem through a systematic molecular modeling and structural bioinformatics approach. Our work shows that simple rigid body rotations may account for the observed collapse of the prepore towards the membrane surface. Support for this idea comes from analysis of published cryo-EM maps of the pneumolysin pore, available crystal structures and molecular dynamics simulations. The latter data in particular reveal that Domains 1, 2 and 4 are able to undergo significant rotational movements with respect to each other. Together, our data provide new and testable insights into the mechanism of pore formation by CDCs.


Asunto(s)
Proteínas Bacterianas/química , Toxinas Bacterianas/química , Membrana Celular/metabolismo , Proteínas Citotóxicas Formadoras de Poros/química , Membrana Celular/química , Colesterol , Simulación de Dinámica Molecular , Conformación Proteica
14.
Immunol Rev ; 235(1): 35-54, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20536554

RESUMEN

The secretory granule-mediated cell death pathway is the key mechanism for elimination of virus-infected and transformed target cells by cytotoxic lymphocytes. The formation of the immunological synapse between an effector and a target cell leads to exocytic trafficking of the secretory granules and the release of their contents, which include pro-apoptotic serine proteases, granzymes, and pore-forming perforin into the synapse. There, perforin polymerizes and forms a transmembrane pore that allows the delivery of granzymes into the cytosol, where they initiate various apoptotic death pathways. Unlike relatively redundant individual granzymes, functional perforin is absolutely essential for cytotoxic lymphocyte function and immune regulation in the host. Nevertheless, perforin is still the least studied and understood cytotoxic molecule in the immune system. In this review, we discuss the current state of affairs in the perforin field: the protein's structure and function as well as its role in immune-mediated diseases.


Asunto(s)
Citotoxicidad Inmunológica , Enfermedades del Sistema Inmune/inmunología , Sinapsis Inmunológicas , Células Asesinas Naturales/inmunología , Perforina/metabolismo , Linfocitos T Citotóxicos/inmunología , Animales , Apoptosis , Granzimas/metabolismo , Humanos , Enfermedades del Sistema Inmune/patología , Modelos Moleculares , Perforina/química , Perforina/genética , Conformación Proteica , Transporte de Proteínas , Vesículas Secretoras/inmunología , Transducción de Señal , Relación Estructura-Actividad
15.
Bioinformatics ; 28(10): 1299-302, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22467914

RESUMEN

MOTIVATION: The ß-barrel is a ubiquitous fold that is deployed to accomplish a wide variety of biological functions including membrane-embedded pores. Key influences of ß-barrel lumen diameter include the number of ß-strands (n) and the degree of shear (S), the latter value measuring the extent to which the ß-sheet is tilted within the ß-barrel. Notably, it has previously been reported that the shear value for small antiparallel ß-barrels (n≤24) typically ranges between n and 2n. Conversely, it has been suggested that the ß-strands in giant antiparallel ß-barrels, such as those formed by pore forming cholesterol-dependent cytolysins (CDC), are parallel relative to the axis of the ß-barrel, i.e. S=0. The S=0 arrangement, however, has never been observed in crystal structures of small ß-barrels. Therefore, the structural basis for how CDCs form a ß-barrel and span a membrane remains to be understood. RESULTS: Through comparison of molecular models with experimental data, we are able to identify how giant CDC ß-barrels utilize a 'near parallel' arrangement of ß-strands where S=n/2. Furthermore, we show how side-chain packing within the ß-barrel lumen is an important limiting factor with respect to the possible shear values for small ß-barrels (n≤24 ß-strands). In contrast, our models reveal no such limitation restricts the shear value of giant ß-barrels (n>24 ß-strands). Giant ß-barrels can thus access a different architecture compared with smaller ß-barrels.


Asunto(s)
Toxinas Bacterianas/química , Proteínas Citotóxicas Formadoras de Poros/química , Estructura Secundaria de Proteína , Proteínas Bacterianas/química , Modelos Moleculares , Estreptolisinas/química
16.
Proc Natl Acad Sci U S A ; 107(12): 5534-9, 2010 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-20212169

RESUMEN

Residues within processed protein fragments bound to major histocompatibility complex class I (MHC-I) glycoproteins have been considered to function as a series of "independent pegs" that either anchor the peptide (p) to the MHC-I and/or interact with the spectrum of alphabeta-T-cell receptors (TCRs) specific for the pMHC-I epitope in question. Mining of the extensive pMHC-I structural database established that many self- and viral peptides show extensive and direct interresidue interactions, an unexpected finding that has led us to the idea of "constrained" peptides. Mutational analysis of two constrained peptides (the HLA B44 restricted self-peptide (B44DPalpha-EEFGRAFSF) and an H2-D(b) restricted influenza peptide (D(b)PA, SSLENFRAYV) demonstrated that the conformation of the prominently exposed arginine in both peptides was governed by interactions with MHC-I-orientated flanking residues from the peptide itself. Using reverse genetics in a murine influenza model, we revealed that mutation of an MHC-I-orientated residue (SSLENFRAYV --> SSLENARAYV) within the constrained PA peptide resulted in a diminished cytotoxic T lymphocyte (CTL) response and the recruitment of a limited pMHC-I specific TCR repertoire. Interactions between individual peptide positions can thus impose fine control on the conformation of pMHC-I epitopes, whereas the perturbation of such constraints can lead to a previously unappreciated mechanism of viral escape.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Linfocitos T/inmunología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Presentación de Antígeno , Epítopos/química , Epítopos/genética , Epítopos/metabolismo , Femenino , Antígenos H-2/química , Antígenos H-2/genética , Antígenos H-2/metabolismo , Antígenos HLA-B/química , Antígenos HLA-B/genética , Antígenos HLA-B/metabolismo , Antígeno HLA-B44 , Antígeno de Histocompatibilidad H-2D , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Conformación Proteica , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/inmunología
17.
Commun Biol ; 6(1): 42, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36639734

RESUMEN

The Membrane Attack Complex (MAC) is responsible for forming large ß-barrel channels in the membranes of pathogens, such as gram-negative bacteria. Off-target MAC assembly on endogenous tissue is associated with inflammatory diseases and cancer. Accordingly, a human C5b-9 specific antibody, aE11, has been developed that detects a neoepitope exposed in C9 when it is incorporated into the C5b-9 complex, but not present in the plasma native C9. For nearly four decades aE11 has been routinely used to study complement, MAC-related inflammation, and pathophysiology. However, the identity of C9 neoepitope remains unknown. Here, we determined the cryo-EM structure of aE11 in complex with polyC9 at 3.2 Å resolution. The aE11 binding site is formed by two separate surfaces of the oligomeric C9 periphery and is therefore a discontinuous quaternary epitope. These surfaces are contributed by portions of the adjacent TSP1, LDLRA, and MACPF domains of two neighbouring C9 protomers. By substituting key antibody interacting residues to the murine orthologue, we validated the unusual binding modality of aE11. Furthermore, aE11 can recognise a partial epitope in purified monomeric C9 in vitro, albeit weakly. Taken together, our results reveal the structural basis for MAC recognition by aE11.


Asunto(s)
Complemento C9 , Complejo de Ataque a Membrana del Sistema Complemento , Humanos , Animales , Ratones , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Complemento C5b , Complemento C9/química , Complemento C9/metabolismo , Proteínas del Sistema Complemento/metabolismo , Epítopos
18.
BMC Evol Biol ; 12: 59, 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22551122

RESUMEN

BACKGROUND: The pore-forming protein perforin is central to the granule-exocytosis pathway used by cytotoxic lymphocytes to kill abnormal cells. Although this mechanism of killing is conserved in bony vertebrates, cytotoxic cells are present in other chordates and invertebrates, and their cytotoxic mechanism has not been elucidated. In order to understand the evolution of this pathway, here we characterize the origins and evolution of perforin. RESULTS: We identified orthologs and homologs of human perforin in all but one species analysed from Euteleostomi, and present evidence for an earlier ortholog in Gnathostomata but not in more primitive chordates. In placental mammals perforin is a single copy gene, but there are multiple perforin genes in all lineages predating marsupials, except birds. Our comparisons of these many-to-one homologs of human perforin show that they mainly arose from lineage-specific gene duplications in multiple taxa, suggesting acquisition of new roles or different modes of regulation. We also present evidence that perforin arose from duplication of the ancient MPEG1 gene, and that it shares a common ancestor with the functionally related complement proteins. CONCLUSIONS: The evolution of perforin in vertebrates involved a complex pattern of gene, as well as intron, gain and loss. The primordial perforin gene arose at least 500 million years ago, at around the time that the major histocompatibility complex-T cell receptor antigen recognition system was established. As it is absent from primitive chordates and invertebrates, cytotoxic cells from these lineages must possess a different effector molecule or cytotoxic mechanism.


Asunto(s)
Evolución Molecular , Duplicación de Gen , Perforina/genética , Vertebrados/genética , Secuencias de Aminoácidos , Animales , Exocitosis , Humanos , Intrones , Filogenia , Alineación de Secuencia , Análisis de Secuencia de Proteína , Linfocitos T Citotóxicos/inmunología , Vertebrados/inmunología
19.
PLoS Pathog ; 6(5): e1000898, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20485572

RESUMEN

Many bacterial pathogens utilize a type III secretion system to deliver multiple effector proteins into host cells. Here we found that the type III effectors, NleE from enteropathogenic E. coli (EPEC) and OspZ from Shigella, blocked translocation of the p65 subunit of the transcription factor, NF-kappaB, to the host cell nucleus. NF-kappaB inhibition by NleE was associated with decreased IL-8 expression in EPEC-infected intestinal epithelial cells. Ectopically expressed NleE also blocked nuclear translocation of p65 and c-Rel, but not p50 or STAT1/2. NleE homologues from other attaching and effacing pathogens as well OspZ from Shigella flexneri 6 and Shigella boydii, also inhibited NF-kappaB activation and p65 nuclear import; however, a truncated form of OspZ from S. flexneri 2a that carries a 36 amino acid deletion at the C-terminus had no inhibitory activity. We determined that the C-termini of NleE and full length OspZ were functionally interchangeable and identified a six amino acid motif, IDSY(M/I)K, that was important for both NleE- and OspZ-mediated inhibition of NF-kappaB activity. We also established that NleB, encoded directly upstream from NleE, suppressed NF-kappaB activation. Whereas NleE inhibited both TNFalpha and IL-1beta stimulated p65 nuclear translocation and IkappaB degradation, NleB inhibited the TNFalpha pathway only. Neither NleE nor NleB inhibited AP-1 activation, suggesting that the modulatory activity of the effectors was specific for NF-kappaB signaling. Overall our data show that EPEC and Shigella have evolved similar T3SS-dependent means to manipulate host inflammatory pathways by interfering with the activation of selected host transcriptional regulators.


Asunto(s)
Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/metabolismo , Shigella boydii/metabolismo , Shigella flexneri/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Virulencia/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Células CACO-2 , Disentería Bacilar/inmunología , Disentería Bacilar/metabolismo , Disentería Bacilar/microbiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/patogenicidad , Células HeLa , Humanos , Proteínas I-kappa B/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Subunidad p50 de NF-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Shigella boydii/patogenicidad , Shigella flexneri/patogenicidad , Activación Transcripcional/fisiología , Virulencia
20.
Proc Natl Acad Sci U S A ; 106(24): 9809-14, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19487666

RESUMEN

The pore-forming protein perforin is critical for defense against many human pathogens and for preventing a catastrophic collapse of immune homeostasis, manifested in infancy as Type 2 familial hemophagocytic lymphohistiocytosis (FHL). However, no evidence has yet linked defective perforin cytotoxicity with cancer susceptibility in humans. Here, we examined perforin function in every patient reported in the literature who lived to at least 10 years of age without developing FHL despite inheriting mutations in both of their perforin (PRF1) alleles. Our analysis showed that almost 50% of these patients developed at least 1 hematological malignancy in childhood or adolescence. The broad range of pathologies argued strongly against a common environmental or viral cause for the extraordinary cancer incidence. Functionally, what distinguished these patients was their inheritance of PRF1 alleles encoding temperature-sensitive missense mutations. By contrast, truly null missense mutations with no rescue at the permissive temperature were associated with the more common severe presentation with FHL in early infancy. Our study provides the first mechanistic evidence for a link between defective perforin-mediated cytotoxicity and cancer susceptibility in humans and establishes the paradigm that temperature sensitivity of perforin function is a predictor of FHL severity.


Asunto(s)
Predisposición Genética a la Enfermedad , Linfohistiocitosis Hemofagocítica/inmunología , Mutación Missense , Perforina/fisiología , Alelos , Humanos , Perforina/química , Perforina/genética , Pliegue de Proteína , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA