Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
EMBO J ; 37(13)2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29858229

RESUMEN

The human gastric pathogen Helicobacter pylori is a major causative agent of gastritis, peptic ulcer disease, and gastric cancer. As part of its adhesive lifestyle, the bacterium targets members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family by the conserved outer membrane adhesin HopQ. The HopQ-CEACAM1 interaction is associated with inflammatory responses and enables the intracellular delivery and phosphorylation of the CagA oncoprotein via a yet unknown mechanism. Here, we generated crystal structures of HopQ isotypes I and II bound to the N-terminal domain of human CEACAM1 (C1ND) and elucidated the structural basis of H. pylori specificity toward human CEACAM receptors. Both HopQ alleles target the ß-strands G, F, and C of C1ND, which form the trans dimerization interface in homo- and heterophilic CEACAM interactions. Using SAXS, we show that the HopQ ectodomain is sufficient to induce C1ND monomerization and thus providing H. pylori a route to influence CEACAM-mediated cell adherence and signaling events.


Asunto(s)
Antígenos CD/fisiología , Proteínas Bacterianas/fisiología , Moléculas de Adhesión Celular/fisiología , Helicobacter pylori/fisiología , Animales , Antígenos CD/química , Proteínas Bacterianas/química , Células CHO , Moléculas de Adhesión Celular/química , Línea Celular Tumoral , Cricetulus , Humanos , Multimerización de Proteína
2.
Mol Microbiol ; 110(1): 33-46, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29995350

RESUMEN

The human stomach pathogen Helicobacter pyloriattaches to healthy and inflamed gastric tissue through members of a paralogous family of 'Helicobacter outer membrane proteins' (Hops), including adhesins BabA, SabA, HopQ, LabA and HopZ. Hops share a conserved 25 kDa C-terminal region that is thought to form an autotransporter-like transmembrane domain. Instead, our results show that Hops contain a non-continuous transmembrane domain, composed of seven predicted ß-strands at the C-terminus and one at the N-terminus. Folding and outer membrane localization of the C-terminal ß-domain critically depends on a predicted transmembrane ß-strand within the first 16 N-terminal residues. The N-terminus is shown to reside in the periplasm, and our crystal and small angle X-ray scattering structures for the SabA extracellular domain reveal a conserved coiled-coil stem domain that connects to transmembrane ß-strand 1 and 2. Taken together, our data show that Hop adhesins represent a novel outer membrane protein topology encompassing an OmpA-like 8-stranded ß-barrel that is interrupted by a 15-108 kDa domain inserted inside the first extracellular loop. The insertion of large, folded domains in an extracellular loop is unprecedented in bacterial outer membrane proteins and is expected to have important consequences on how these proteins reach the cell surface.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Helicobacter pylori/fisiología , Sistemas de Secreción Tipo V/metabolismo , Adhesinas Bacterianas/química , Adhesinas Bacterianas/genética , Simulación por Computador , Helicobacter pylori/genética , Filogenia , Conformación Proteica en Lámina beta/genética , Dominios Proteicos/genética , Transporte de Proteínas/fisiología , Dispersión del Ángulo Pequeño , Análisis de Secuencia de Proteína , Eliminación de Secuencia , Sistemas de Secreción Tipo V/química
3.
J Virol ; 90(1): 611-5, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26468526

RESUMEN

We report the crystal structure of the M2 ectodomain (M2e) in complex with a monoclonal antibody that binds the amino terminus of M2. M2e extends into the antibody binding site to form an N-terminal ß-turn near the bottom of the paratope. This M2e folding differs significantly from that of M2e in complex with an antibody that binds another part of M2e. This suggests that M2e can adopt at least two conformations that can elicit protective antibodies.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Antivirales/química , Proteínas de la Matriz Viral/química , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/metabolismo , Anticuerpos Antivirales/aislamiento & purificación , Anticuerpos Antivirales/metabolismo , Línea Celular , Cristalografía por Rayos X , Humanos , Ratones Endogámicos BALB C , Unión Proteica , Conformación Proteica , Proteínas de la Matriz Viral/metabolismo
4.
Vet Res ; 48(1): 86, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29216932

RESUMEN

Campylobacter infections are among the most prevalent foodborne infections in humans, resulting in a massive disease burden worldwide. Broilers have been identified as the major source of campylobacteriosis and reducing Campylobacter loads in the broiler caeca has been proposed as an effective measure to decrease the number of infections in humans. Failure of current methods to control Campylobacter in broilers stresses the urgency to develop novel mitigation measures. We obtained six nanobodies with a broad specificity, that recognize strains belonging to the two most relevant species, Campylobacter jejuni and Campylobacter coli. The target of the nanobodies was identified as the major outer membrane protein, a porin that contributes to bacterial virulence and viability. Multimerization of the nanobodies led to agglutination of C. jejuni cells, which may affect colonization in the chicken gut. These Campylobacter-specific nanobodies may be useful to develop a strategy for preserving chickens from Campylobacter colonization.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Infecciones por Campylobacter/veterinaria , Campylobacter coli/inmunología , Campylobacter jejuni/inmunología , Pollos , Enfermedades de las Aves de Corral/prevención & control , Anticuerpos de Dominio Único/inmunología , Animales , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Bacterianas/inmunología , Infecciones por Campylobacter/inmunología , Infecciones por Campylobacter/microbiología , Infecciones por Campylobacter/prevención & control , Epítopos/inmunología , Porinas/inmunología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/microbiología
5.
J Biol Chem ; 290(13): 8409-19, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25631050

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) strains are important causes of intestinal disease in humans and lead to severe production losses in animal farming. A range of fimbrial adhesins in ETEC strains determines host and tissue tropism. ETEC strains expressing F4 fimbriae are associated with neonatal and post-weaning diarrhea in piglets. Three naturally occurring variants of F4 fimbriae (F4ab, F4ac, and F4ad) exist that differ in the primary sequence of their major adhesive subunit FaeG, and each features a related yet distinct receptor binding profile. Here the x-ray structure of FaeGad bound to lactose provides the first structural insight into the receptor specificity and mode of binding by the poly-adhesive F4 fimbriae. A small D'-D″-α1-α2 subdomain grafted on the immunoglobulin-like core of FaeG hosts the carbohydrate binding site. Two short amino acid stretches Phe(150)-Glu(152) and Val(166)-Glu(170) of FaeGad bind the terminal galactose in the lactosyl unit and provide affinity and specificity to the interaction. A hemagglutination-based assay with E. coli expressing mutant F4ad fimbriae confirmed the elucidated co-complex structure. Interestingly, the crucial D'-α1 loop that borders the FaeGad binding site adopts a different conformation in the two other FaeG variants and hints at a heterogeneous binding pocket among the FaeG serotypes.


Asunto(s)
Adhesinas de Escherichia coli/química , Antígenos Bacterianos/química , Escherichia coli Enterotoxigénica/metabolismo , Proteínas de Escherichia coli/química , Proteínas Fimbrias/química , Secuencia de Aminoácidos , Antígenos Bacterianos/metabolismo , Antígenos CD/química , Sitios de Unión , Cristalografía por Rayos X , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Galactosilceramidas/química , Lactosa/química , Lactosilceramidos/química , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica
6.
J Biol Chem ; 289(36): 25374-81, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25053417

RESUMEN

Shiga toxin Stx2e is the major known agent that causes edema disease in newly weaned pigs. This severe disease is characterized by neurological disorders, hemorrhagic lesions, and frequent fatal outcomes. Stx2e consists of an enzymatically active A subunit and five B subunits that bind to a specific glycolipid receptor on host cells. It is evident that antibodies binding to the A subunit or the B subunits of Shiga toxin variants may have the capability to inhibit their cytotoxicity. Here, we report the discovery and characterization of a VHH single domain antibody (nanobody) isolated from a llama phage display library that confers potent neutralizing capacity against Stx2e toxin. We further present the crystal structure of the complex formed between the nanobody (NbStx2e1) and the Stx2e toxoid, determined at 2.8 Å resolution. Structural analysis revealed that for each B subunit of Stx2e, one NbStx2e1 is interacting in a head-to-head orientation and directly competing with the glycolipid receptor binding site on the surface of the B subunit. The neutralizing NbStx2e1 can in the future be used to prevent or treat edema disease.


Asunto(s)
Anticuerpos Neutralizantes/química , Estructura Terciaria de Proteína , Toxina Shiga II/química , Anticuerpos de Dominio Único/química , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/inmunología , Sitios de Unión/genética , Sitios de Unión/inmunología , Unión Competitiva/inmunología , Camélidos del Nuevo Mundo/inmunología , Cristalografía por Rayos X , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica/inmunología , Subunidades de Proteína/química , Subunidades de Proteína/inmunología , Subunidades de Proteína/metabolismo , Receptores de Superficie Celular/metabolismo , Homología de Secuencia de Aminoácido , Toxina Shiga II/inmunología , Toxina Shiga II/metabolismo , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología
7.
Vet Res ; 46: 14, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25828907

RESUMEN

Enterotoxigenic Escherichia coli that cause neonatal and post-weaning diarrhea in piglets express F4 fimbriae to mediate attachment towards host receptors. Recently we described how llama single domain antibodies (VHHs) fused to IgA, produced in Arabidopsis thaliana seeds and fed to piglets resulted in a progressive decline in shedding of F4 positive ETEC bacteria. Here we present the structures of these inhibiting VHHs in complex with the major adhesive subunit FaeG. A conserved surface, distant from the lactose binding pocket, is targeted by these VHHs, highlighting the possibility of targeting epitopes on single-domain adhesins that are non-involved in receptor binding.


Asunto(s)
Adhesinas de Escherichia coli/inmunología , Diarrea/veterinaria , Escherichia coli Enterotoxigénica/fisiología , Infecciones por Escherichia coli/veterinaria , Fimbrias Bacterianas/inmunología , Anticuerpos de Dominio Único/química , Enfermedades de los Porcinos/inmunología , Animales , Camélidos del Nuevo Mundo/inmunología , Cristalografía por Rayos X/veterinaria , Diarrea/inmunología , Diarrea/microbiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Anticuerpos de Dominio Único/inmunología , Porcinos , Enfermedades de los Porcinos/microbiología , Esparcimiento de Virus
8.
Trop Anim Health Prod ; 47(1): 117-22, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25311441

RESUMEN

Post-weaning diarrhea (PWD) caused by enterotoxigenic Escherichia coli (ETEC) is an important disease of newly weaned piglets. ETEC strains commonly express F4 and/or F18 fimbriae that attach to carbohydrate receptors present on the intestinal epithelium during colonization. The disease status in the Ugandan piggeries had previously not been studied. In this cross-sectional sero-survey and clinical outbreak monitoring, we found very high sero-prevalence levels of both anti-F4 (70.5%) and anti-F18 (73.7%) antibodies, despite limited cases of clinical outbreaks. Strains isolated from these cases were typically F18(+) ETEC. High antibiotic resistance and multi-drug resistance were characteristics of the isolates, with highest resistance level of over 95% to commonly used antibiotics such as penicillin and tetracycline. We conclude that ETEC infections are widely spread on farms in Central Uganda but clinical disease outbreaks were masked by the management practices on these farms, like the use of extensive antibiotic prophylaxis.


Asunto(s)
Profilaxis Antibiótica/métodos , Diarrea/veterinaria , Escherichia coli Enterotoxigénica , Infecciones por Escherichia coli/veterinaria , Enfermedades de los Porcinos/epidemiología , Animales , Antibacterianos/farmacología , Estudios Transversales , Diarrea/microbiología , Brotes de Enfermedades , Ensayo de Inmunoadsorción Enzimática , Proteínas de Escherichia coli/inmunología , Proteínas Fimbrias/inmunología , Fimbrias Bacterianas , Pruebas de Sensibilidad Microbiana , Reacción en Cadena de la Polimerasa , Prevalencia , Estudios Seroepidemiológicos , Porcinos/microbiología , Enfermedades de los Porcinos/microbiología , Uganda/epidemiología , Destete
9.
Mol Microbiol ; 90(2): 267-77, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23927005

RESUMEN

Binary fission is the ultimate step of the prokaryotic cell cycle. In Gram-negative bacteria like Escherichia coli, this step implies the invagination of three biological layers (cytoplasmic membrane, peptidoglycan and outer membrane), biosynthesis of the new poles and eventually, daughter cells separation. The latter requires the coordinated action of the N-acetylmuramyl-L-alanine amidases AmiA/B/C and their LytM activators EnvC and NlpD to cleave the septal peptidoglycan. We present here the 2.5 Å crystal structure of AmiC which includes the first report of an AMIN domain structure, a ß-sandwich of two symmetrical four-stranded ß-sheets exposing highly conserved motifs on the two outer faces. We show that this N-terminal domain, involved in the localization of AmiC at the division site, is a new peptidoglycan-binding domain. The C-terminal catalytic domain shows an auto-inhibitory alpha helix obstructing the active site. AmiC lacking this helix exhibits by itself an activity comparable to that of the wild type AmiC activated by NlpD. We also demonstrate the interaction between AmiC and NlpD by microscale thermophoresis and confirm the importance of the active site blocking alpha helix in the regulation of the amidase activity.


Asunto(s)
División Celular , Escherichia coli/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/química , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Peptidoglicano/metabolismo , Secuencias de Aminoácidos , Proteínas Bacterianas/metabolismo , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Lipoproteínas/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/genética , Conformación Proteica , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
10.
Mol Microbiol ; 86(1): 82-95, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22812428

RESUMEN

F18-positive enterotoxigenic and Shiga toxin-producing Escherichia coli are responsible for post-weaning diarrhoea and oedema disease in pigs and lead to severe production losses in the farming industry. F18 fimbriae attach to the small intestine of young piglets by latching onto glycosphingolipids with A/H blood group determinants on type 1 core. We demonstrate the N-terminal domain of the F18 fimbrial subunit FedF to be responsible for ABH-mediated attachment and present its X-ray structure in ligand-free form and bound to A and B type 1 hexaoses. The FedF lectin domain comprises a 10-stranded immunoglobulin-like ß-sandwich. Three linear motives, Q(47) -N(50), H(88) -S(90) and R(117) -T(119), form a shallow glycan binding pocket near the tip of the domain that is selective for type 1 core glycans in extended conformation. In addition to the glycan binding pocket, a polybasic loop on the membrane proximal surface of FedF lectin domain is shown to be required for binding to piglet enterocytes. Although dispensable for ABH glycan recognition, the polybasic surface adds binding affinity in the context of the host cell membrane, a mechanism that is proposed to direct ABH-glycan binding to cell-bound glycosphingolipids and could allow bacteria to avoid clearance by secreted glycoproteins.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/química , Sistema del Grupo Sanguíneo ABO/metabolismo , Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/química , Animales , Sitios de Unión , Línea Celular , Cristalografía por Rayos X , Enterocitos/microbiología , Modelos Moleculares , Unión Proteica , Conformación Proteica , Porcinos
11.
bioRxiv ; 2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37292721

RESUMEN

The majority of the world population carry the gastric pathogen Helicobacter pylori. Fortunately, most individuals experience only low-grade or no symptoms, but in many cases the chronic inflammatory infection develops into severe gastric disease, including duodenal ulcer disease and gastric cancer. Here we report on a protective mechanism where H. pylori attachment and accompanying chronic mucosal inflammation can be reduced by antibodies that are present in a vast majority of H. pylori carriers. These antibodies block binding of the H. pylori attachment protein BabA by mimicking BabA's binding to the ABO blood group glycans in the gastric mucosa. However, many individuals demonstrate low titers of BabA blocking antibodies, which is associated with an increased risk for duodenal ulceration, suggesting a role for these antibodies in preventing gastric disease.

12.
J Neurosci ; 31(43): 15320-8, 2011 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22031878

RESUMEN

Mutations in the small heat shock protein HSPB1 (HSP27) are causative for Charcot-Marie-Tooth (CMT) neuropathy. We previously showed that a subset of these mutations displays higher chaperone activity and enhanced affinity to client proteins. We hypothesized that this excessive binding property might cause the HSPB1 mutant proteins to disturb the function of proteins essential for the maintenance or survival of peripheral neurons. In the present work, we explored this hypothesis further and compared the protein complexes formed by wild-type and mutant HSPB1. Tubulin came out as the most striking differential interacting protein, with hyperactive mutants binding more strongly to both tubulin and microtubules. This anomalous binding leads to a stabilization of the microtubule network in a microtubule-associated protein-like manner as reflected by resistance to cold depolymerization, faster network recovery after nocodazole treatment, and decreased rescue and catastrophe rates of individual microtubules. In a transgenic mouse model for mutant HSPB1 that recapitulates all features of CMT, we could confirm the enhanced interaction of mutant HSPB1 with tubulin. Increased stability of the microtubule network was also clear in neurons isolated from these mice. Since neuronal cells are particularly vulnerable to disturbances in microtubule dynamics, this mechanism might explain the neuron-specific CMT phenotype caused by HSPB1 mutations.


Asunto(s)
Proteínas de Choque Térmico HSP27/genética , Microtúbulos/metabolismo , Mutación/genética , Neuronas/metabolismo , Análisis de Varianza , Animales , Células Cultivadas , Chlorocebus aethiops , Ganglios Espinales/citología , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas de Choque Térmico , Humanos , Hielo/efectos adversos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Chaperonas Moleculares , Neuronas/efectos de los fármacos , Nocodazol/farmacología , Unión Proteica , Resonancia por Plasmón de Superficie , Espectrometría de Masas en Tándem/métodos , Factores de Tiempo , Transfección/métodos , Tubulina (Proteína)/genética , Tubulina (Proteína)/farmacología , Moduladores de Tubulina/farmacología
13.
Microorganisms ; 8(4)2020 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-32218315

RESUMEN

Attachment to the host gastric mucosa is a key step in Helicobacter pylori infection. Recently, a novel adhesin, HopQ, was shown to bind distinct host CEACAM proteins-an interaction that was found to be essential for the translocation of CagA, a key virulence factor of H. pylori. The HopQ-CEACAM1 co-crystal structure revealed a binding mode dependent on loops in HopQ that are clasped by disulfide bonds. In this study, we investigated the importance of these cysteine residues for CEACAM1 engagement by H. pylori. We observed a loss of CEACAM1 binding and CagA translocation upon disruption of the disulfide bond in loop CL1 (connecting C103 to C132 in HopQ). Deletion of the Dsb-like oxidoreductase HP0231 did not affect cell surface expression of HopQ or alter the interaction of H. pylori with target cells. Although HP0231 deletion was previously described to impede CagA translocation, our results indicate that this occurs through a HopQ-independent mechanism. Together, our results open up new avenues to therapeutically target the HopQ-CEACAM1 interaction and reduce the burden of pathogenic H. pylori.

14.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 5): 411-20, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19390146

RESUMEN

Many Gram-negative bacteria use the chaperone-usher pathway to express adhesive surface structures, such as fimbriae, in order to mediate attachment to host cells. Periplasmic chaperones are required to shuttle fimbrial subunits or pilins through the periplasmic space in an assembly-competent form. The chaperones cap the hydrophobic surface of the pilins through a donor-strand complementation mechanism. FaeE is the periplasmic chaperone required for the assembly of the F4 fimbriae of enterotoxigenic Escherichia coli. The FaeE crystal structure shows a dimer formed by interaction between the pilin-binding interfaces of the two monomers. Dimerization and tetramerization have been observed previously in crystal structures of fimbrial chaperones and have been suggested to serve as a self-capping mechanism that protects the pilin-interactive surfaces in solution in the absence of the pilins. However, thermodynamic and biochemical data show that FaeE occurs as a stable monomer in solution. Other lines of evidence indicate that self-capping of the pilin-interactive interfaces is not a mechanism that is conservedly applied by all periplasmic chaperones, but is rather a case-specific solution to cap aggregation-prone surfaces.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/química , Chaperonas Moleculares/química , Adhesinas de Escherichia coli/química , Rastreo Diferencial de Calorimetría , Reactivos de Enlaces Cruzados/farmacología , Cristalografía por Rayos X , Dimerización , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/aislamiento & purificación , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Glutaral/farmacología , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/aislamiento & purificación , Chaperonas Moleculares/metabolismo , Nefelometría y Turbidimetría , Conformación Proteica , Desnaturalización Proteica , Mapeo de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación
15.
Curr Opin Struct Biol ; 44: 48-58, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28043017

RESUMEN

Infectious disease processes like bacterial adherence or the activity of secreted toxins frequently gain host and tissue specificity by glycan binding interactions with the host glycome. Recent functional and structural studies highlight the high niche specialization of bacterial lectins, but also reveal a remarkable plasticity in their glycan binding sites and mechanisms, to adapt to host glycome dynamics or changing environmental conditions at the site of infection. In this review we put emphasis on new structural insights in host adaptation and dynamics of bacterial carbohydrate binding adhesins and toxins in human pathogens like uropathogenic and enteropathogenic Escherichia coli, Helicobacter pylori, Yersinia pestis or Vibrio cholerae. Also, structure-aided drug design to counteract glycan-mediated host-pathogen interactions is coming of age, with the design of novel anti-adhesive compounds and (single-domain) antibodies that target glycan binding sites.


Asunto(s)
Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Evolución Molecular , Polisacáridos/metabolismo , Adaptación Fisiológica , Animales , Adhesión Bacteriana , Humanos
16.
Cell Host Microbe ; 21(3): 376-389, 2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28279347

RESUMEN

The BabA adhesin mediates high-affinity binding of Helicobacter pylori to the ABO blood group antigen-glycosylated gastric mucosa. Here we show that BabA is acid responsive-binding is reduced at low pH and restored by acid neutralization. Acid responsiveness differs among strains; often correlates with different intragastric regions and evolves during chronic infection and disease progression; and depends on pH sensor sequences in BabA and on pH reversible formation of high-affinity binding BabA multimers. We propose that BabA's extraordinary reversible acid responsiveness enables tight mucosal bacterial adherence while also allowing an effective escape from epithelial cells and mucus that are shed into the acidic bactericidal lumen and that bio-selection and changes in BabA binding properties through mutation and recombination with babA-related genes are selected by differences among individuals and by changes in gastric acidity over time. These processes generate diverse H. pylori subpopulations, in which BabA's adaptive evolution contributes to H. pylori persistence and overt gastric disease.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Concentración de Iones de Hidrógeno
17.
Cell Host Microbe ; 19(1): 55-66, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26764597

RESUMEN

The Helicobacter pylori adhesin BabA binds mucosal ABO/Le(b) blood group (bg) carbohydrates. BabA facilitates bacterial attachment to gastric surfaces, increasing strain virulence and forming a recognized risk factor for peptic ulcers and gastric cancer. High sequence variation causes BabA functional diversity, but the underlying structural-molecular determinants are unknown. We generated X-ray structures of representative BabA isoforms that reveal a polymorphic, three-pronged Le(b) binding site. Two diversity loops, DL1 and DL2, provide adaptive control to binding affinity, notably ABO versus O bg preference. H. pylori strains can switch bg preference with single DL1 amino acid substitutions, and can coexpress functionally divergent BabA isoforms. The anchor point for receptor binding is the embrace of an ABO fucose residue by a disulfide-clasped loop, which is inactivated by reduction. Treatment with the redox-active pharmaceutic N-acetylcysteine lowers gastric mucosal neutrophil infiltration in H. pylori-infected Le(b)-expressing mice, providing perspectives on possible H. pylori eradication therapies.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/química , Sistema del Grupo Sanguíneo ABO/metabolismo , Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Polisacáridos/metabolismo , Sistema del Grupo Sanguíneo ABO/genética , Adhesinas Bacterianas/genética , Animales , Sitios de Unión , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Helicobacter pylori/química , Helicobacter pylori/genética , Humanos , Ratones , Modelos Moleculares , Unión Proteica
18.
Nat Microbiol ; 2: 16189, 2016 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-27748768

RESUMEN

Helicobacter pylori specifically colonizes the human gastric epithelium and is the major causative agent for ulcer disease and gastric cancer development. Here, we identify members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family as receptors of H. pylori and show that HopQ is the surface-exposed adhesin that specifically binds human CEACAM1, CEACAM3, CEACAM5 and CEACAM6. HopQ-CEACAM binding is glycan-independent and targeted to the N-domain. H. pylori binding induces CEACAM1-mediated signalling, and the HopQ-CEACAM1 interaction enables translocation of the virulence factor CagA into host cells and enhances the release of pro-inflammatory mediators such as interleukin-8. Based on the crystal structure of HopQ, we found that a ß-hairpin insertion (HopQ-ID) in HopQ's extracellular 3+4 helix bundle domain is important for CEACAM binding. A peptide derived from this domain competitively inhibits HopQ-mediated activation of the Cag virulence pathway, as genetic or antibody-mediated abrogation of the HopQ function shows. Together, our data suggest the HopQ-CEACAM1 interaction to be a potentially promising novel therapeutic target to combat H. pylori-associated diseases.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Moléculas de Adhesión Celular/metabolismo , Helicobacter pylori/fisiología , Helicobacter pylori/patogenicidad , Interacciones Huésped-Patógeno , Adhesinas Bacterianas/química , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Línea Celular , Cristalografía por Rayos X , Humanos , Interleucina-8/metabolismo , Unión Proteica , Conformación Proteica , Transporte de Proteínas , Virulencia
19.
PLoS One ; 9(12): e114691, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25502211

RESUMEN

Post-weaning diarrhea and edema disease caused by F18 fimbriated E. coli are important diseases in newly weaned piglets and lead to severe production losses in farming industry. Protective treatments against these infections have thus far limited efficacy. In this study we generated nanobodies directed against the lectin domain of the F18 fimbrial adhesin FedF and showed in an in vitro adherence assay that four unique nanobodies inhibit the attachment of F18 fimbriated E. coli bacteria to piglet enterocytes. Crystallization of the FedF lectin domain with the most potent inhibitory nanobodies revealed their mechanism of action. These either competed with the binding of the blood group antigen receptor on the FedF surface or induced a conformational change in which the CDR3 region of the nanobody displaces the D″-E loop adjacent to the binding site. This D″-E loop was previously shown to be required for the interaction between F18 fimbriated bacteria and blood group antigen receptors in a membrane context. This work demonstrates the feasibility of inhibiting the attachment of fimbriated pathogens by employing nanobodies directed against the adhesin domain.


Asunto(s)
Adhesión Bacteriana/inmunología , Escherichia coli/citología , Escherichia coli/fisiología , Fimbrias Bacterianas/metabolismo , Anticuerpos de Dominio Único/inmunología , Adhesinas Bacterianas/química , Adhesinas Bacterianas/inmunología , Adhesinas Bacterianas/metabolismo , Animales , Unión Competitiva , Camélidos del Nuevo Mundo/inmunología , Camélidos del Nuevo Mundo/microbiología , Metabolismo de los Hidratos de Carbono , Enterocitos/microbiología , Escherichia coli/inmunología , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/inmunología , Proteínas de Escherichia coli/metabolismo , Fimbrias Bacterianas/inmunología , Expresión Génica , Modelos Moleculares , Conformación Proteica , Anticuerpos de Dominio Único/genética , Porcinos/inmunología , Porcinos/microbiología
20.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 12): 1631-5, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25484214

RESUMEN

Helicobacter pylori is a human pathogen that colonizes about 50% of the world's population, causing chronic gastritis, duodenal ulcers and even gastric cancer. A steady emergence of multiple antibiotic resistant strains poses an important public health threat and there is an urgent requirement for alternative therapeutics. The blood group antigen-binding adhesin BabA mediates the intimate attachment to the host mucosa and forms a major candidate for novel vaccine and drug development. Here, the recombinant expression and crystallization of a soluble BabA truncation (BabA(25-460)) corresponding to the predicted extracellular adhesin domain of the protein are reported. X-ray diffraction data for nanobody-stabilized BabA(25-460) were collected to 2.25 Šresolution from a crystal that belonged to space group P21, with unit-cell parameters a = 50.96, b = 131.41, c = 123.40 Å, α = 90.0, ß = 94.8, γ = 90.0°, and which was predicted to contain two BabA(25-460)-nanobody complexes per asymmetric unit.


Asunto(s)
Adhesinas Bacterianas/química , Antígenos de Grupos Sanguíneos/inmunología , Helicobacter pylori/inmunología , Adhesinas Bacterianas/aislamiento & purificación , Secuencia de Bases , Cristalografía por Rayos X , Cartilla de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA