Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Infect Immun ; 83(4): 1372-83, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25605771

RESUMEN

Infections by Neisseria gonorrhoeae are increasingly common, are often caused by antibiotic-resistant strains, and can result in serious and lasting sequelae, prompting the reemergence of gonococcal disease as a leading global health concern. N. gonorrhoeae is a human-restricted pathogen that primarily colonizes urogenital mucosal surfaces. Disease progression varies greatly between the sexes: men usually present with symptomatic infection characterized by a painful purulent urethral discharge, while in women, the infection is often asymptomatic, with the most severe pathology occurring when the bacteria ascend from the lower genital tract into the uterus and fallopian tubes. Classical clinical studies demonstrated that clinically infectious strains uniformly express Opa adhesins; however, their specificities were unknown at the time. While in vitro studies have since identified CEACAM proteins as the primary target of Opa proteins, the gonococcal specificity for this human family of receptors has not been addressed in the context of natural infection. In this study, we characterize a collection of low-passage-number clinical-specimen-derived N. gonorrhoeae isolates for Opa expression and assess their CEACAM-binding profiles. We report marked in vivo selection for expression of phase-variable Opa proteins that bind CEACAM1 and CEACAM5 but selection against expression of Opa variants that bind to the neutrophil-restricted decoy receptor CEACAM3. This is the first study showing phenotypic selection for distinct CEACAM-binding phenotypes in vivo, and it supports the opposing functions of CEACAMs that facilitate infection versus driving inflammation within the genital tract.


Asunto(s)
Antígenos CD/metabolismo , Adhesión Bacteriana/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Gonorrea/inmunología , Adhesinas Bacterianas/metabolismo , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Degranulación de la Célula/inmunología , Línea Celular , Cuello del Útero/microbiología , Femenino , Proteínas Ligadas a GPI/metabolismo , Gonorrea/microbiología , Humanos , Inflamación/inmunología , Masculino , Neisseria gonorrhoeae/inmunología , Neisseria gonorrhoeae/aislamiento & purificación , Neutrófilos/inmunología , Unión Proteica , Isoformas de Proteínas/metabolismo , Uretra/microbiología
2.
Proc Natl Acad Sci U S A ; 108(37): 15174-8, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21876142

RESUMEN

Bacterial cell surfaces are commonly decorated with a layer formed from multiple copies of adhesin proteins whose binding interactions initiate colonization and infection processes. In this study, we investigate the physical deformability of the UspA1 adhesin protein from Moraxella catarrhalis, a causative agent of middle-ear infections in humans. UspA1 binds a range of extracellular proteins including fibronectin, and the epithelial cellular receptor carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1). Electron microscopy indicates that unliganded UspA1 is densely packed at, and extends about 800 Å from, the Moraxella surface. Using a modified atomic force microscope, we show that the adhesive properties and thickness of the UspA1 layer at the cell surface varies on addition of either fibronectin or CEACAM1. This in situ analysis is then correlated with the molecular structure of UspA1. To provide an overall model for UspA1, we have determined crystal structures for two N-terminal fragments which are then combined with a previous structure of the CEACAM1-binding site. We show that the UspA1-fibronectin complex is formed between UspA1 head region and the 13th type-III domain of fibronectin and, using X-ray scattering, that the complex involves an angular association between these two proteins. In combination with a previous study, which showed that the CEACAM1-UspA1 complex is distinctively bent in solution, we correlate these observations on isolated fragments of UspA1 with its in situ response on the cell surface. This study therefore provides a rare direct demonstration of protein conformational change at the cell surface.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Antígenos CD/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Moléculas de Adhesión Celular/metabolismo , Fibronectinas/metabolismo , Mecanotransducción Celular , Moraxella catarrhalis/metabolismo , Adhesinas Bacterianas/ultraestructura , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/ultraestructura , Membrana Celular/metabolismo , Humanos , Microscopía de Fuerza Atómica , Modelos Moleculares , Moraxella catarrhalis/ultraestructura , Unión Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X
3.
Cell Microbiol ; 14(9): 1402-14, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22519722

RESUMEN

Interactions between Mycobacterium tuberculosis bacilli and alveolar macrophages have been extensively characterized, while similar analyses in epithelial cells have not been performed. In this study, we microscopically examined endosomal trafficking of M. tuberculosis strain Erdman in A549 cells, a human type II pneumocyte cell line. Immuno-electron microscopic (IEM) analyses indicate that M. tuberculosis bacilli are internalized to a compartment labelled first with Rab5 and then with Rab7 small GTPase proteins. This suggests that, unlike macrophages, M. tuberculosis bacilli traffic to late endosomes in epithelial cells. However, fusion of lysosomes with the bacteria-containing compartment appears to be inhibited, as illustrated by IEM studies employing LAMP-2 and cathepsin-L antibodies. Examination by transmission electron microscopy and IEM revealed M. tuberculosis-containing compartments surrounded by double membranes and labelled with antibodies against the autophagy marker Lc3, providing evidence for involvement and intersection of the autophagy and endosomal pathways. Interestingly, inhibition of the autophagy pathway using 3-methyladenine improved host cell viability and decreased numbers of viable intracellular bacteria recovered after 72 h post infection. Collectively, these data suggest that trafficking patterns for M. tuberculosis bacilli in alveolar epithelial cells differ from macrophages, and that autophagy is involved this process.


Asunto(s)
Autofagia , Células Epiteliales/microbiología , Células Epiteliales/fisiología , Mycobacterium tuberculosis/patogenicidad , Línea Celular , Endosomas/microbiología , Células Epiteliales/ultraestructura , Humanos , Lisosomas/metabolismo , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica
4.
Mol Microbiol ; 82(5): 1129-49, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22050461

RESUMEN

Complement evasion is an important survival strategy of Neisseria meningitidis (Nm) during colonization and infection. Previously, we have shown that Nm Opc binds to serum vitronectin to inhibit complement-mediated killing. In this study, we demonstrate meningococcal interactions with vitronectin via a novel adhesin, Msf (meningococcal surface fibril, previously NhhA or Hsf). As with Opc, Msf binds preferentially to activated vitronectin (aVn), engaging at its N-terminal region but the C-terminal heparin binding domain may also participate. However, unlike Opc, the latter binding is not heparin-mediated. By binding to aVn, Msf or Opc can impart serum resistance, which is further increased in coexpressers, a phenomenon dependent on serum aVn concentrations. The survival fitness of aVn-binding derivatives was evident from mixed population studies, in which msf/opc mutants were preferentially depleted. In addition, using vitronectin peptides to block Msf-aVn interactions, aVn-induced inhibition of lytic C5b-9 formation and of serum killing could be reversed. As Msf-encoding gene is ubiquitous in the meningococcal strains examined and is expressed in vivo, serum resistance via Msf may be of significance to meningococcal pathogenesis. The data imply that vitronectin binding may be an important strategy for the in vivo survival of Nm for which the bacterium has evolved redundant mechanisms.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Actividad Bactericida de la Sangre , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Interacciones Huésped-Patógeno , Neisseria meningitidis/inmunología , Neisseria meningitidis/patogenicidad , Vitronectina/metabolismo , Evasión Inmune , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Neisseria meningitidis/metabolismo , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas
5.
EMBO J ; 27(12): 1779-89, 2008 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-18497748

RESUMEN

Moraxella catarrhalis is a ubiquitous human-specific bacterium commonly associated with upper and lower respiratory tract infections, including otitis media, sinusitis and chronic obstructive pulmonary disease. The bacterium uses an autotransporter protein UspA1 to target an important human cellular receptor carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1). Using X-ray crystallography, we show that the CEACAM1 receptor-binding region of UspA1 unusually consists of an extended, rod-like left-handed trimeric coiled-coil. Mutagenesis and binding studies of UspA1 and the N-domain of CEACAM1 have been used to delineate the interacting surfaces between ligand and receptor and guide assembly of the complex. However, solution scattering, molecular modelling and electron microscopy analyses all indicate that significant bending of the UspA1 coiled-coil stalk also occurs. This explains how UspA1 can engage CEACAM1 at a site far distant from its head group, permitting closer proximity of the respective cell surfaces during infection.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Antígenos CD/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Moléculas de Adhesión Celular/metabolismo , Adhesinas Bacterianas/química , Antígenos CD/química , Proteínas de la Membrana Bacteriana Externa/química , Sitios de Unión , Moléculas de Adhesión Celular/química , Dicroismo Circular , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Secundaria de Proteína , Receptores de Superficie Celular , Termodinámica
6.
PLoS Pathog ; 6(5): e1000911, 2010 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-20502634

RESUMEN

The host vasculature is believed to constitute the principal route of dissemination of Neisseria meningitidis (Nm) throughout the body, resulting in septicaemia and meningitis in susceptible humans. In vitro, the Nm outer membrane protein Opc can enhance cellular entry and exit, utilising serum factors to anchor to endothelial integrins; but the mechanisms of binding to serum factors are poorly characterised. This study demonstrates that Nm Opc expressed in acapsulate as well as capsulate bacteria can increase human brain endothelial cell line (HBMEC) adhesion and entry by first binding to serum vitronectin and, to a lesser extent, fibronectin. This study also demonstrates that Opc binds preferentially to the activated form of human vitronectin, but not to native vitronectin unless the latter is treated to relax its closed conformation. The direct binding of vitronectin occurs at its Connecting Region (CR) requiring sulphated tyrosines Y(56) and Y(59). Accordingly, Opc/vitronectin interaction could be inhibited with a conformation-dependent monoclonal antibody 8E6 that targets the sulphotyrosines, and with synthetic sulphated (but not phosphorylated or unmodified) peptides spanning the vitronectin residues 43-68. Most importantly, the 26-mer sulphated peptide bearing the cell-binding domain (45)RGD(47) was sufficient for efficient meningococcal invasion of HBMECs. To our knowledge, this is the first study describing the binding of a bacterial adhesin to sulphated tyrosines of the host receptor. Our data also show that a single region of Opc is likely to interact with the sulphated regions of both vitronectin and of heparin. As such, in the absence of heparin, Opc-expressing Nm interact directly at the CR but when precoated with heparin, they bind via heparin to the heparin-binding domain of the activated vitronectin, although with a lower affinity than at the CR. Such redundancy suggests the importance of Opc/vitronectin interaction in meningococcal pathogenesis and may enable the bacterium to harness the benefits of the physiological processes in which the host effector molecule participates.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Encéfalo/citología , Células Endoteliales/microbiología , Neisseria meningitidis Serogrupo A/metabolismo , Neisseria meningitidis Serogrupo B/metabolismo , Vitronectina/metabolismo , Animales , Adhesión Bacteriana/fisiología , Proteínas de la Membrana Bacteriana Externa/genética , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/microbiología , Bovinos , Línea Celular , Células Endoteliales/citología , Fibronectinas/metabolismo , Heparina/química , Heparina/metabolismo , Humanos , Ratones , Neisseria meningitidis Serogrupo A/genética , Neisseria meningitidis Serogrupo B/genética , Fosforilación/fisiología , Desnaturalización Proteica , Estructura Terciaria de Proteína , Especificidad de la Especie , Sulfatos/metabolismo , Tirosina/metabolismo , Vitronectina/química
7.
Clin Sci (Lond) ; 118(9): 547-64, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20132098

RESUMEN

The human species is the only natural host of Neisseria meningitidis, an important cause of bacterial meningitis globally, and, despite its association with devastating diseases, N. meningitidis is a commensal organism found frequently in the respiratory tract of healthy individuals. To date, antibiotic resistance is relatively uncommon in N. meningitidis isolates but, due to the rapid onset of disease in susceptible hosts, the mortality rate remains approx. 10%. Additionally, patients who survive meningococcal disease often endure numerous debilitating sequelae. N. meningitidis strains are classified primarily into serogroups based on the type of polysaccharide capsule expressed. In total, 13 serogroups have been described; however, the majority of disease is caused by strains belonging to one of only five serogroups. Although vaccines have been developed against some of these, a universal meningococcal vaccine remains a challenge due to successful immune evasion strategies of the organism, including mimicry of host structures as well as frequent antigenic variation. N. meningitidis express a range of virulence factors including capsular polysaccharide, lipopolysaccharide and a number of surface-expressed adhesive proteins. Variation of these surface structures is necessary for meningococci to evade killing by host defence mechanisms. Nonetheless, adhesion to host cells and tissues needs to be maintained to enable colonization and ensure bacterial survival in the niche. The aims of the present review are to provide a brief outline of meningococcal carriage, disease and burden to society. With this background, we discuss several bacterial strategies that may enable its survival in the human respiratory tract during colonization and in the blood during infection. We also examine several known meningococcal adhesion mechanisms and conclude with a section on the potential processes that may operate in vivo as meningococci progress from the respiratory niche through the blood to reach the central nervous system.


Asunto(s)
Infecciones Meningocócicas/microbiología , Neisseria meningitidis/patogenicidad , Portador Sano , Interacciones Huésped-Patógeno , Humanos , Meninges/microbiología , Infecciones Meningocócicas/terapia , Nasofaringe/microbiología , Neisseria meningitidis/clasificación , Neisseria meningitidis/fisiología , Virulencia
8.
Cell Microbiol ; 11(3): 389-405, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19016781

RESUMEN

Neisseria meningitidis Opc protein is an effective invasin for human endothelial cells. We have investigated novel human endothelial receptors targeted by Opc and observed that Opc-expressing bacteria interacted with a 100 kDa protein in whole-cell lysates of human endothelial and epithelial cells. The identity of the protein was established as alpha-actinin by mass spectrometry. Opc expression was essential for the recognition of alpha-actinin whether provided in a purified form or in cell extracts. The interaction of the two proteins did not involve intermediate molecules. As there was no demonstrable expression of alpha-actinin on the surfaces of any of the eight cell lines studied, the likelihood of the interactions after meningococcal internalization was examined. Confocal imaging demonstrated considerable colocalization of N. meningitidis with alpha-actinin especially after a prolonged period of internalization. This may imply that bacteria and alpha-actinin initially occur in separate compartments and co-compartmentalization occurs progressively over the 8 h infection period used. In conclusion, these studies have identified a novel and an intracellular target for the N. meningitidis Opc invasin. Since alpha-actinin is a modulator of a variety of signalling pathways and of cytoskeletal functions, its targeting by Opc may enable bacteria to survive/translocate across endothelial barriers.


Asunto(s)
Actinina/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas del Citoesqueleto/metabolismo , Neisseria meningitidis/fisiología , Línea Celular , Células Cultivadas , Células Endoteliales/microbiología , Células Epiteliales , Humanos , Espectrometría de Masas , Microscopía Confocal , Unión Proteica
9.
Infect Immun ; 77(11): 5170-80, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19720754

RESUMEN

T cells may interact with a number of bacterial surface antigens, an encounter which has the potential to downmodulate host immune responses. Neisseria meningitidis, a human colonizer and an agent of septicemia and meningitis, expresses Opa proteins which interact with the CEACAM1 receptor expressed on activated T cells. Since CEACAM1 can act as an inhibitory receptor and T cells in subepithelial tissues may encounter whole bacteria, which often express Opa proteins in vivo, this study assessed primarily if Opa proteins expressed on meningococci affect T-cell functions. In addition, Opa-containing outer membrane vesicles (OMV) have been used as vaccine antigens, and therefore Opa+ and Opa- OMV were also studied. While Opa+ bacteria adhered to CEACAM-expressing T cells, both the Opa+ and Opa- phenotypes induced no to a small transient depression, followed by a prolonged increase in proliferation as well as cytokine production. Such responses were also observed with heat-killed bacteria or OMV. In addition, while anti-CEACAM antibodies alone inhibited proliferation, on coincubation of T cells with bacteria and the antibodies, bacterial effects predominated and were Opa independent. Thus, while Opa proteins of N. meningitidis can bind to T-cell-expressed CEACAM1, this is not sufficient to overcome the T-cell recognition of bacterial factors, which results in a proliferative and cytokine response, an observation consistent with the ability of the host to establish lasting immunity to Opa-expressing meningococci that it frequently encounters. The data also imply that Opa-proficient vaccine preparations may not necessarily inhibit T-cell functions via CEACAM1 binding.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Linfocitos T CD4-Positivos/microbiología , Infecciones Meningocócicas/inmunología , Neisseria gonorrhoeae/inmunología , Neisseria meningitidis/inmunología , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Antígenos CD/inmunología , Antígenos CD/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Activación de Linfocitos/inmunología , Infecciones Meningocócicas/metabolismo , Neisseria gonorrhoeae/metabolismo , Neisseria meningitidis/metabolismo , Fenotipo
10.
Pediatr Infect Dis J ; 28(1): 43-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19057458

RESUMEN

Nontypeable Haemophilus influenzae is a significant pathogen in children, causing otitis media, sinusitis, conjunctivitis, pneumonia, and occasionally invasive infections. H. influenzae type b conjugate vaccines have no effect on infections caused by nontypeable strains because nontypeable strains are nonencapsulated. Approximately, one-third of episodes of otitis media are caused by nontypeable H. influenzae and the bacterium is the most common cause of recurrent otitis media. Recent progress in elucidating molecular mechanisms of pathogenesis, understanding the role of biofilms in otitis media and an increasing understanding of immune responses have potential for development of novel strategies to improve prevention and treatment of otitis media caused by nontypeable H. influenzae. Feasibility of vaccination for prevention of otitis media due to nontypeable H. influenzae was recently demonstrated in a clinical trial with a vaccine that included the surface virulence factor, protein D.


Asunto(s)
Infecciones por Haemophilus/microbiología , Haemophilus influenzae/patogenicidad , Animales , Portador Sano/microbiología , Fibrosis Quística/microbiología , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/patología , Infecciones por Haemophilus/prevención & control , Vacunas contra Haemophilus/inmunología , Vacunas contra Haemophilus/uso terapéutico , Haemophilus influenzae/clasificación , Haemophilus influenzae/aislamiento & purificación , Humanos , Nasofaringe/microbiología , Otitis Media/microbiología
11.
Top Curr Chem ; 288: 139-56, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-22328029

RESUMEN

Microbial adhesion is generally a complex process, involving multiple adhesins on a single microbe and their respective target receptors on host cells. In some situations, various adhesins of a microbe may co-operate in an apparently hierarchical and sequential manner whereby the first adhesive event triggers the target cell to express receptors for additional microbial adhesins. In other instances, adhesins may act in concert leading to high avidity interactions, often a prelude to cellular invasion and tissue penetration. Mechanisms used to target the host include both lectin-like interactions and protein-protein interactions; the latter are often highly specific for the host or a tissue within the host. This reflective chapter aims to offer a point of view on microbial adhesion by presenting some experiences and thoughts especially related to respiratory pathogens and explore if there can be any future hope of controlling bacterial infections via preventing adhesion or invasion stages of microbial pathogenesis.

12.
J Oral Microbiol ; 11(1): 1565043, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30719234

RESUMEN

Neisseria meningitidis, Haemophilus influenzae, and Moraxella catarrhalis are pathogenic bacteria adapted to reside on human respiratory mucosal epithelia. One common feature of these species is their ability to target members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family, especially CEACAM1, which is achieved via structurally distinct ligands expressed by each species. Beside respiratory epithelial cells, cells at the dentogingival junction express high levels of CEACAM1. It is possible that bacterial species resident within the oral cavity also utilise CEACAM1 for colonisation and invasion of gingival tissues. From a screen of 59 isolates from the human oral cavity representing 49 bacterial species, we identified strains from Fusobacterium bound to CEACAM1. Of the Fusobacterium species tested, the CEACAM1-binding property was exhibited by F. nucleatum (Fn) and F. vincentii (Fv) but not F. polymorphum (Fp) or F. animalis (Fa) strains tested. These studies identified that CEACAM adhesion was mediated using a trimeric autotransporter adhesin (TAA) for which no function has thus far been defined. We therefore propose the name CEACAM binding protein of Fusobacterium (CbpF). CbpF was identified to be present in the majority of unspeciated Fusobacterium isolates confirming a subset of Fusobacterium spp. are able to target human CEACAM1.

13.
Inflamm Bowel Dis ; 14(2): 162-75, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17941093

RESUMEN

BACKGROUND: Mucosally adherent E. coli are found in inflammatory bowel disease (IBD) and colon cancer. They promote release of the proinflammatory cytokine interleukin-8 (IL-8). We explored mechanisms for this release and its inhibition by drugs. METHODS: IL-8 release from colon epithelial cells in response to mucosal E. coli isolates from IBD, colon cancer, and controls was characterized at the cellular and molecular level. RESULTS: IL-8 response of HT29 cells was greater with Crohn's disease (689 +/- 298 [mean +/- SD] pg IL-8/mL at 4 hours, n = 7) and colon cancer isolates (532 +/- 415 pg/mL, n = 14) than with ulcerative colitis (236 +/- 58 pg/mL, n = 6) or control isolates (236 +/- 100 pg/mL, n = 6, P < 0.0001). Bacterial supernatants contained shed flagellin that triggered IL-8 release. For whole bacteria the IL-8 response to E. coli that agglutinate red blood cells (548 +/- 428 pg IL-8/mL, n = 16), a function that correlates with epithelial invasion, was greater than for nonhemagglutinators (281 +/- 253 pg/mL, n = 17; P < 0.0001). This was particularly marked among E. coli that, although flagellate, could not release IL-8 from TLR5-transfected HEK293 cells. IL-8 release was mediated by extracellular-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) and inhibited by mesalamine, but not hydrocortisone, at therapeutic concentrations. CONCLUSIONS: Mucosa-associated E. coli shed flagellin that elicits epithelial IL-8 release but this may only become relevant when the mucosal barrier is weakened to expose basolateral TLR5. Adherent and invasive IBD and colon cancer E. coli isolates also elicit a flagellin-independent IL-8 response that may be relevant when the mucosal barrier is intact. The IL-8 release is MAPK-dependent and inhibited by mesalamine.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Escherichia coli/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Interleucina-8/antagonistas & inhibidores , Interleucina-8/metabolismo , Mesalamina/farmacología , Estudios de Casos y Controles , Células Cultivadas , Neoplasias del Colon/inmunología , Neoplasias del Colon/microbiología , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Flagelina/genética , Flagelina/inmunología , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Sistema de Señalización de MAP Quinasas
14.
PLoS One ; 13(3): e0193940, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29547646

RESUMEN

Neisseria meningitidis is an antigenically and genetically variable Gram-negative bacterium and a causative agent of meningococcal meningitis and septicaemia. Meningococci encode many outer membrane proteins, including Opa, Opc, Msf, fHbp and NadA, identified as being involved in colonisation of the host and evasion of the immune response. Although vaccines are available for the prevention of some types of meningococcal disease, none currently offer universal protection. We have used sequences within the Neisseria PubMLST database to determine the variability of msf and opc in 6,500 isolates. In-silico analysis revealed that although opc is highly conserved, it is not present in all isolates, with most isolates in clonal complex ST-11 lacking a functional opc. In comparison, msf is found in all meningococcal isolates, and displays diversity in the N-terminal domain. We identified 20 distinct Msf sequence variants (Msf SV), associated with differences in number of residues within the putative Vn binding motifs. Moreover, we showed distinct correlations with certain Msf SVs and isolates associated with either hyperinvasive lineages or those clonal complexes associated with a carriage state. We have demonstrated differences in Vn binding between three Msf SVs and generated a cross reactive Msf polyclonal antibody. Our study has highlighted the importance of using large datasets to inform vaccine development and provide further information on the antigenic diversity exhibited by N. meningitidis.


Asunto(s)
Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/genética , Vacunas Meningococicas/genética , Neisseria meningitidis/genética , Adhesinas Bacterianas/genética , Secuencia de Aminoácidos , Variación Antigénica/genética , Biología Computacional/métodos , Variación Genética/genética , Humanos , Meningitis Meningocócica/inmunología , Alineación de Secuencia
15.
PLoS One ; 10(3): e0124133, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25826209

RESUMEN

The human pathogen Neisseria meningitides (Nm) attains serum resistance via a number of mechanisms, one of which involves binding to the host complement regulator protein vitronectin. We have shown previously that the Meningococcal surface fibril (Msf), a trimeric autotransporter, binds to the activated form of vitronectin (aVn) to increase Nm survival in human serum. In this study, we aimed to identify the aVn-binding region of Msf to assess its potential as an antigen which can elicit antibodies that block aVn binding and/or possess bactericidal properties. Using several recombinant Msf fragments spanning its surface-exposed region, the smallest aVn-binding recombinants were found to span residues 1-86 and 39-124. The use of further deletion constructs and overlapping recombinant Msf fragments suggested that a region of Msf comprising residues 39-82 may be primarily important for aVn binding and that other regions may also be involved but to a lesser extent. Molecular modelling implicated K66 and K68, conserved in all available Msf sequences, to be involved in the interaction. Recombinant fragments which bound to aVn were able to reduce the survival advantage conveyed by aVn-interaction in serum bactericidal assays. Antibodies raised against one such fragment inhibited aVn binding to Msf. In addition, the antibodies enhanced specific killing of Msf-expressing Nm in a dose-dependent manner. Overall, this study identifies an aVn-binding region of Msf, an adhesin known to impart serum resistance properties to the pathogen; and shows that this region of Msf can elicit antibodies with dual properties which reduce pathogen survival within the host and thus has potential as a vaccine antigen.


Asunto(s)
Proteínas Bacterianas/metabolismo , Neisseria meningitidis/metabolismo , Vitronectina/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Datos de Secuencia Molecular
16.
PLoS One ; 9(3): e90999, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24599281

RESUMEN

Circulating monocytes in the bloodstream typically migrate to other tissues and differentiate into tissue resident macrophages, the process being determined by the constituents of the microenvironments encountered. These may include microbes and their products. In this study, we investigated whether Moraxella catarrhalis Ubiquitous Surface Protein A1 (UspA1), known to bind to a widely expressed human cell surface receptor CEACAM1, influences monocyte differentiation as receptor engagement has been shown to have profound effects on monocytes. We used the recombinant molecules corresponding to the regions of UspA1 which either bind (rD-7; UspA1527-665) or do not bind (r6-8; UspA1659-863) to CEACAM1 and investigated their effects on CD206, CD80 and CD86 expression on freshly isolated human CD14+ monocytes from peripheral blood mononuclear cells (PBMC). Exposure to rD-7, but not r6-8, biased monocyte differentiation towards a CD14+CD206+ phenotype, with reduced CD80 expression. Monocytes treated with rD-7 also secreted high levels of IL-1ra and chemokine IL-8 but not IL-10 or IL-12p70. The effects of rD-7 were independent of any residual endotoxin. Unexpectedly, these effects of rD-7 were also independent of its ability to bind to CEACAM1, as monocyte pre-treatment with the anti-CEACAM antibody A0115 known to inhibit rD-7 binding to the receptor, did not affect rD-7-driven differentiation. Further, another control protein rD-7/D (a mutant form of rD-7, known not to bind to CEACAMs), also behaved as the parent molecule. Our data suggest that specific regions of M. catarrhalis adhesin UspA1 may modulate inflammation during infection through a yet unknown receptor on monocytes.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Diferenciación Celular/efectos de los fármacos , Monocitos/citología , Moraxella catarrhalis/metabolismo , Proteínas Recombinantes/farmacología , Anticuerpos Antibacterianos/metabolismo , Antígenos CD/metabolismo , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Quimiocinas/metabolismo , Fluorescencia , Humanos , Lectinas Tipo C/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Fenotipo , Receptores de Superficie Celular/metabolismo , Reproducibilidad de los Resultados
17.
Methods Mol Biol ; 799: 143-52, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21993644

RESUMEN

Meningococcal mechanisms of adhesion are complex, involving multiple adhesins and their respective target receptors on host cells. Three major surface structures--pili, Opa, and Opc--have been known for some time to mediate meningococcal adhesion to target human cells. More recently, several other relatively minor adhesins have also come to light. The literature on bacterial adhesion mechanisms provides numerous examples of various adhesins acting cooperatively in an apparently hierarchical and sequential manner; in other instances, adhesins may act in concert leading to high avidity interactions, often a prelude to cellular invasion and tissue penetration. Such examples are also present in the case of meningococci, although our knowledge of adhesin cooperation and synergy is far from complete. Meningococcal mechanisms used to target the host, which are often specific for the host or a tissue within the host, include both lectin-like interactions and protein-protein interactions; the latter tend to determine specificity in general. Understanding (a) what determines specificity (i.e. molecular features of adhesins and receptors), (b) encourages cellular penetration (i.e. adhesin pairs, which act in concert or synergistically to deliver effective signals for invasion and induce other cellular responses), (c) level of redundancy (more than one mechanisms of targeting host receptors), (d) host situations that encourage tissue penetration (inflammatory situations during which circulating cytokines upregulate target cell receptors, effectively encouraging greater adhesion/invasion), and (e) down-stream effects on host functions in general are all clearly important in our future strategies of controlling meningococcal pathogenesis.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana/fisiología , Moléculas de Adhesión Celular/metabolismo , Interacciones Huésped-Patógeno , Neisseria meningitidis/metabolismo , Receptores de Superficie Celular/metabolismo , Fimbrias Bacterianas/metabolismo , Humanos , Neisseria meningitidis/fisiología , Unión Proteica , Receptores Toll-Like/metabolismo
18.
PLoS One ; 7(9): e45452, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23049802

RESUMEN

Moraxella catarrhalis (Mx) is a common cause of otitis media and exacerbation of chronic obstructive pulmonary disease, an increasing worldwide problem. Surface proteins UspA1 and UspA2 of Mx bind to a number of human receptors and may function in pathogenesis. Genetic recombination events in the pathogen can generate hybrid proteins termed UspA2H. However, whether certain key functions (e.g. UspA1-specific CEACAM binding) can be exchanged between these adhesin families remains unknown. In this study, we have shown that Mx can incorporate the UspA1 CEACAM1-binding region not only into rare UspA1 proteins devoid of CEACAM-binding ability, but also into UspA2 which normally lack this capacity. Further, a screen of Mx isolates revealed the presence of novel UspA2 Variant proteins (UspA2V) in ∼14% of the CEACAM-binding population. We demonstrate that the expression of UspA2/2V with the CEACAM-binding domain enable Mx to bind both to cell surface CEACAMs and to integrins, the latter via vitronectin. Such properties of UspA2/2V have not been reported to date. The studies demonstrate that the UspA family is much more heterogeneous than previously believed and illustrate the in vivo potential for exchange of functional regions between UspA proteins which could convey novel adhesive functions whilst enhancing immune evasion.


Asunto(s)
Antígenos CD/genética , Proteínas de la Membrana Bacteriana Externa/genética , Moléculas de Adhesión Celular/genética , Moraxella catarrhalis/genética , Proteínas Mutantes Quiméricas/genética , Secuencia de Aminoácidos , Antígenos CD/metabolismo , Proteínas de la Membrana Bacteriana Externa/clasificación , Proteínas de la Membrana Bacteriana Externa/aislamiento & purificación , Proteínas de la Membrana Bacteriana Externa/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Humanos , Integrinas/genética , Integrinas/metabolismo , Datos de Secuencia Molecular , Moraxella catarrhalis/aislamiento & purificación , Moraxella catarrhalis/metabolismo , Infecciones por Moraxellaceae/microbiología , Proteínas Mutantes Quiméricas/aislamiento & purificación , Proteínas Mutantes Quiméricas/metabolismo , Otitis Media/microbiología , Filogenia , Unión Proteica , Estructura Terciaria de Proteína , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Recombinación Genética , Alineación de Secuencia , Transformación Bacteriana , Vitronectina/genética , Vitronectina/metabolismo
19.
J Vis Exp ; (44)2010 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-21085092

RESUMEN

The Opc protein of Neisseria meningitidis (Nm, meningococcus) is a surface-expressed integral outer membrane protein, which can act as an adhesin and an effective invasin for human epithelial and endothelial cells. We have identified endothelial surface-located integrins as major receptors for Opc, a process which requires Opc to first bind to integrin ligands such as vitronectin and via these to the cell-expressed receptors(1). This process leads to bacterial invasion of endothelial cells(2). More recently, we observed an interaction of Opc with a 100 kDa protein found in whole cell lysates of human cells(3). We initially observed this interaction when host cell proteins separated by electrophoresis and blotted on to nitrocellulose were overlaid with Opc-expressing Nm. The interaction was direct and did not involve intermediate molecules. By mass spectrometry, we established the identity of the protein as α-actinin. As no surface expressed α-actinin was found on any of the eight cell lines examined, and as Opc interactions with endothelial cells in the presence of serum lead to bacterial entry into the target cells, we examined the possibility of the two proteins interacting intracellularly. For this, cultured human brain microvascular endothelial cells (HBMECs) were infected with Opc-expressing Nm for extended periods and the locations of internalised bacteria and α-actinin were examined by confocal microscopy. We observed time-dependent increase in colocalisation of Nm with the cytoskeletal protein, which was considerable after an eight hour period of bacterial internalisation. In addition, the use of quantitative imaging software enabled us to obtain a relative measure of the colocalisation of Nm with α-actinin and other cytoskeletal proteins. Here we present a protocol for visualisation and quantification of the colocalisation of the bacterium with intracellular proteins after bacterial entry into human endothelial cells, although the procedure is also applicable to human epithelial cells.


Asunto(s)
Actinina/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Células Endoteliales/microbiología , Microscopía Confocal/métodos , Neisseria meningitidis/metabolismo , Encéfalo/irrigación sanguínea , Células Endoteliales/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA