Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 47(3): 510-523.e4, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28930661

RESUMEN

Within the interleukin 1 (IL-1) cytokine family, IL-1 receptor accessory protein (IL-1RAcP) is the co-receptor for eight receptor-cytokine pairs, including those involving cytokines IL-1ß and IL-33. Unlike IL-1ß, IL-33 does not have a signaling complex that includes both its cognate receptor, ST2, and the shared co-receptor IL-1RAcP, which we now present here. Although the IL-1ß and IL-33 complexes shared structural features and engaged identical molecular surfaces of IL-1RAcP, these cytokines had starkly different strategies for co-receptor engagement and signal activation. Our data suggest that IL-1ß binds to IL-1RI to properly present the cytokine to IL-1RAcP, whereas IL-33 binds to ST2 in order to conformationally constrain the cognate receptor in an IL-1RAcP-receptive state. These findings indicate that members of the IL-1 family of cytokines use distinct molecular mechanisms to signal through their shared co-receptor, and they provide the foundation from which to design new therapies to target IL-33 signaling.


Asunto(s)
Interleucina-1/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Animales , Sitios de Unión , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Interleucina-1/química , Proteína 1 Similar al Receptor de Interleucina-1/química , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/química , Interleucina-33/metabolismo , Ratones , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Mutación , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores de Interleucina-1/química , Receptores de Interleucina-1/genética
2.
Proc Natl Acad Sci U S A ; 120(20): e2208673120, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37155900

RESUMEN

The immune deficiency (IMD) pathway directs host defense in arthropods upon bacterial infection. In Pancrustacea, peptidoglycan recognition proteins sense microbial moieties and initiate nuclear factor-κB-driven immune responses. Proteins that elicit the IMD pathway in non-insect arthropods remain elusive. Here, we show that an Ixodes scapularis homolog of croquemort (Crq), a CD36-like protein, promotes activation of the tick IMD pathway. Crq exhibits plasma membrane localization and binds the lipid agonist 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol. Crq regulates the IMD and jun N-terminal kinase signaling cascades and limits the acquisition of the Lyme disease spirochete B. burgdorferi. Additionally, nymphs silenced for crq display impaired feeding and delayed molting to adulthood due to a deficiency in ecdysteroid synthesis. Collectively, we establish a distinct mechanism for arthropod immunity outside of insects and crustaceans.


Asunto(s)
Artrópodos , Infecciones Bacterianas , Borrelia burgdorferi , Ixodes , Enfermedad de Lyme , Animales , Ixodes/microbiología , Borrelia burgdorferi/genética , FN-kappa B , Enfermedad de Lyme/microbiología
3.
EMBO J ; 40(7): e106103, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33522633

RESUMEN

Streptococcus agalactiae, also known as group B Streptococcus (GBS), is the major cause of neonatal sepsis in humans. A critical step to infection is adhesion of bacteria to epithelial surfaces. GBS adhesins have been identified to bind extracellular matrix components and cellular receptors. However, several putative adhesins have no host binding partner characterised. We report here that surface-expressed ß protein of GBS binds to human CEACAM1 and CEACAM5 receptors. A crystal structure of the complex showed that an IgSF domain in ß represents a novel Ig-fold subtype called IgI3, in which unique features allow binding to CEACAM1. Bioinformatic assessment revealed that this newly identified IgI3 fold is not exclusively present in GBS but is predicted to be present in adhesins from other clinically important human pathogens. In agreement with this prediction, we found that CEACAM1 binds to an IgI3 domain found in an adhesin from a different streptococcal species. Overall, our results indicate that the IgI3 fold could provide a broadly applied mechanism for bacteria to target CEACAMs.


Asunto(s)
Adhesinas Bacterianas/química , Antígenos CD/química , Antígeno Carcinoembrionario/química , Moléculas de Adhesión Celular/química , Adhesinas Bacterianas/metabolismo , Animales , Antígenos CD/metabolismo , Sitios de Unión , Células CHO , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Cricetinae , Cricetulus , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/metabolismo , Células HeLa , Humanos , Unión Proteica , Streptococcus agalactiae/metabolismo
4.
PLoS Pathog ; 19(5): e1010979, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37253071

RESUMEN

In its simplest form, bacterial flagellar filaments are composed of flagellin proteins with just two helical inner domains, which together comprise the filament core. Although this minimal filament is sufficient to provide motility in many flagellated bacteria, most bacteria produce flagella composed of flagellin proteins with one or more outer domains arranged in a variety of supramolecular architectures radiating from the inner core. Flagellin outer domains are known to be involved in adhesion, proteolysis and immune evasion but have not been thought to be required for motility. Here we show that in the Pseudomonas aeruginosa PAO1 strain, a bacterium that forms a ridged filament with a dimerization of its flagellin outer domains, motility is categorically dependent on these flagellin outer domains. Moreover, a comprehensive network of intermolecular interactions connecting the inner domains to the outer domains, the outer domains to one another, and the outer domains back to the inner domain filament core, is required for motility. This inter-domain connectivity confers PAO1 flagella with increased stability, essential for its motility in viscous environments. Additionally, we find that such ridged flagellar filaments are not unique to Pseudomonas but are, instead, present throughout diverse bacterial phyla.


Asunto(s)
Bacterias , Flagelina , Flagelina/metabolismo , Bacterias/metabolismo , Flagelos/metabolismo , Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo
5.
Nat Chem Biol ; 19(2): 218-229, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36443572

RESUMEN

Bifidobacteria are early colonizers of the human gut and play central roles in human health and metabolism. To thrive in this competitive niche, these bacteria evolved the capacity to use complex carbohydrates, including mammalian N-glycans. Herein, we elucidated pivotal biochemical steps involved in high-mannose N-glycan utilization by Bifidobacterium longum. After N-glycan release by an endo-ß-N-acetylglucosaminidase, the mannosyl arms are trimmed by the cooperative action of three functionally distinct glycoside hydrolase 38 (GH38) α-mannosidases and a specific GH125 α-1,6-mannosidase. High-resolution cryo-electron microscopy structures revealed that bifidobacterial GH38 α-mannosidases form homotetramers, with the N-terminal jelly roll domain contributing to substrate selectivity. Additionally, an α-glucosidase enables the processing of monoglucosylated N-glycans. Notably, the main degradation product, mannose, is isomerized into fructose before phosphorylation, an unconventional metabolic route connecting it to the bifid shunt pathway. These findings shed light on key molecular mechanisms used by bifidobacteria to use high-mannose N-glycans, a perennial carbon and energy source in the intestinal lumen.


Asunto(s)
Bifidobacterium longum , Manosa , Animales , Humanos , Manosa/metabolismo , Bifidobacterium longum/metabolismo , Microscopía por Crioelectrón , Polisacáridos/química , Manosidasas/metabolismo , Glicósido Hidrolasas/química , Bifidobacterium/metabolismo , Mamíferos
6.
Nat Immunol ; 12(1): 54-61, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21131964

RESUMEN

The mechanisms of HLA-DM-catalyzed peptide exchange remain uncertain. Here we found that all stages of the interaction of HLA-DM with HLA-DR were dependent on the occupancy state of the peptide-binding groove. High-affinity peptides were protected from removal by HLA-DM through two mechanisms: peptide binding induced the dissociation of a long-lived complex of empty HLA-DR and HLA-DM, and high-affinity HLA-DR-peptide complexes bound HLA-DM only very slowly. Nonbinding covalent HLA-DR-peptide complexes were converted into efficient HLA-DM binders after truncation of an N-terminal peptide segment that emptied the P1 pocket and disrupted conserved hydrogen bonds to HLA-DR. HLA-DM thus binds only to HLA-DR conformers in which a critical part of the binding site is already vacant because of spontaneous peptide motion.


Asunto(s)
Antígenos HLA-D/metabolismo , Antígeno HLA-DR2/metabolismo , Proteínas Mutantes/metabolismo , Fragmentos de Péptidos/metabolismo , Animales , Presentación de Antígeno , Células CHO , Catálisis , Cricetinae , Cricetulus , Antígenos HLA-D/química , Antígenos HLA-D/genética , Antígeno HLA-DR2/química , Antígeno HLA-DR2/genética , Humanos , Modelos Químicos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Unión Proteica , Resonancia por Plasmón de Superficie , Transgenes/genética
7.
J Biol Chem ; 297(2): 101011, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34324829

RESUMEN

N-glycosylation is one of the most abundant posttranslational modifications of proteins, essential for many physiological processes, including protein folding, protein stability, oligomerization and aggregation, and molecular recognition events. Defects in the N-glycosylation pathway cause diseases that are classified as congenital disorders of glycosylation. The ability to manipulate protein N-glycosylation is critical not only to our fundamental understanding of biology but also for the development of new drugs for a wide range of human diseases. Chemoenzymatic synthesis using engineered endo-ß-N-acetylglucosaminidases (ENGases) has been used extensively to modulate the chemistry of N-glycosylated proteins. However, defining the molecular mechanisms by which ENGases specifically recognize and process N-glycans remains a major challenge. Here we present the X-ray crystal structure of the ENGase EndoBT-3987 from Bacteroides thetaiotaomicron in complex with a hybrid-type glycan product. In combination with alanine scanning mutagenesis, molecular docking calculations and enzymatic activity measurements conducted on a chemically engineered monoclonal antibody substrate unveil two mechanisms for hybrid-type recognition and processing by paradigmatic ENGases. Altogether, the experimental data provide pivotal insight into the molecular mechanism of substrate recognition and specificity for GH18 ENGases and further advance our understanding of chemoenzymatic synthesis and remodeling of homogeneous N-glycan glycoproteins.


Asunto(s)
Bacteroides thetaiotaomicron/enzimología , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/metabolismo , Simulación del Acoplamiento Molecular/métodos , Polisacáridos/metabolismo , Elementos Estructurales de las Proteínas , Bacteroides thetaiotaomicron/química , Cristalografía por Rayos X , Glicosilación , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/química , Especificidad por Sustrato
8.
EMBO J ; 37(13)2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29724755

RESUMEN

Helicobacter pylori infects half of the world's population, and strains that encode the cag type IV secretion system for injection of the oncoprotein CagA into host gastric epithelial cells are associated with elevated levels of cancer. CagA translocation into host cells is dependent on interactions between the H. pylori adhesin protein HopQ and human CEACAMs. Here, we present high-resolution structures of several HopQ-CEACAM complexes and CEACAMs in their monomeric and dimeric forms establishing that HopQ uses a coupled folding and binding mechanism to engage the canonical CEACAM dimerization interface for CEACAM recognition. By combining mutagenesis with biophysical and functional analyses, we show that the modes of CEACAM recognition by HopQ and CEACAMs themselves are starkly different. Our data describe precise molecular mechanisms by which microbes exploit host CEACAMs for infection and enable future development of novel oncoprotein translocation inhibitors and H. pylori-specific antimicrobial agents.


Asunto(s)
Antígenos Bacterianos/fisiología , Antígenos CD/fisiología , Proteínas Bacterianas/fisiología , Moléculas de Adhesión Celular/fisiología , Helicobacter pylori/fisiología , Proteínas Oncogénicas/fisiología , Antígenos CD/química , Proteínas Bacterianas/química , Moléculas de Adhesión Celular/química , Células HEK293 , Humanos , Mutagénesis , Multimerización de Proteína , Transporte de Proteínas
9.
PLoS Comput Biol ; 17(7): e1009103, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34310592

RESUMEN

Antibodies bind foreign antigens with high affinity and specificity leading to their neutralization and/or clearance by the immune system. The conserved N-glycan on IgG has significant impact on antibody effector function, with the endoglycosidases of Streptococcus pyogenes deglycosylating the IgG to evade the immune system, a process catalyzed by the endoglycosidase EndoS2. Studies have shown that two of the four domains of EndoS2, the carbohydrate binding module (CBM) and the glycoside hydrolase (GH) domain are critical for catalytic activity. To yield structural insights into contributions of the CBM and the GH domains as well as the overall flexibility of EndoS2 to the proteins' catalytic activity, models of EndoS2-Fc complexes were generated through enhanced-sampling molecular-dynamics (MD) simulations and site-identification by ligand competitive saturation (SILCS) docking followed by reconstruction and multi-microsecond MD simulations. Modeling results predict that EndoS2 initially interacts with the IgG through its CBM followed by interactions with the GH yielding catalytically competent states. These may involve the CBM and GH of EndoS2 simultaneously interacting with either the same Fc CH2/CH3 domain or individually with the two Fc CH2/CH3 domains, with EndoS2 predicted to assume closed conformations in the former case and open conformations in the latter. Apo EndoS2 is predicted to sample both the open and closed states, suggesting that either complex can directly form following initial IgG-EndoS2 encounter. Interactions of the CBM and GH domains with the IgG are predicted to occur through both its glycan and protein regions. Simulations also predict that the Fc glycan can directly transfer from the CBM to the GH, facilitating formation of catalytically competent complexes and how the 734 to 751 loop on the CBM can facilitate extraction of the glycan away from the Fc CH2/CH3 domain. The predicted models are compared and consistent with Hydrogen/Deuterium Exchange data. In addition, the complex models are consistent with the high specificity of EndoS2 for the glycans on IgG supporting the validity of the predicted models.


Asunto(s)
Proteínas Bacterianas , Glicósido Hidrolasas , Fragmentos Fc de Inmunoglobulinas , Inmunoglobulina G , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Biología Computacional , Medición de Intercambio de Deuterio , Glicósido Hidrolasas/química , Glicósido Hidrolasas/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Simulación de Dinámica Molecular , Polisacáridos/química , Polisacáridos/metabolismo , Conformación Proteica , Streptococcus pyogenes/enzimología , Especificidad por Sustrato
10.
J Biol Chem ; 294(1): 218-230, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30413536

RESUMEN

Type IV pili (T4P) are bacterial appendages composed of protein subunits, called pilins, noncovalently assembled into helical fibers. T4P are essential, in many bacterial species, for processes as diverse as twitching motility, natural competence, biofilm or microcolony formation, and host cell adhesion. The genes encoding type IV pili are found universally in the Gram-negative, aerobic, nonflagellated, and pathogenic coccobacillus Acinetobacter baumannii, but there is considerable variation in PilA, the major protein subunit, both in amino acid sequence and in glycosylation patterns. Here we report the X-ray crystal structure of PilA from AB5075, a recently characterized, highly virulent isolate, at 1.9 Å resolution and compare it to homologues from A. baumannii strains ACICU and BIDMC57, which are C-terminally glycosylated. These structural comparisons revealed that PilAAB5075 exhibits a distinctly electronegative surface chemistry. To understand the functional consequences of this change in surface electrostatics, we complemented a ΔpilA knockout strain with divergent pilA genes from ACICU, BIDMC57, and AB5075. The resulting transgenic strains showed differential twitching motility and biofilm formation while maintaining the ability to adhere to epithelial cells. PilAAB5075 and PilAACICU, although structurally similar, promote different characteristics, favoring twitching motility and biofilm formation, respectively. These results support a model in which differences in pilus electrostatics affect the equilibrium of microcolony formation, which in turn alters the balance between motility and biofilm formation in Acinetobacter.


Asunto(s)
Acinetobacter baumannii/química , Proteínas Fimbrias/química , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Sustitución de Aminoácidos , Cristalografía por Rayos X , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Glicosilación , Mutación Missense , Dominios Proteicos
11.
Glycobiology ; 30(4): 268-279, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-31172182

RESUMEN

The conserved N-glycan on Asn297 of immunoglobulin G (IgG) has significant impacts on antibody effector functions, and is a frequent target for antibody engineering. Chemoenzymatic synthesis has emerged as a strategy for producing antibodies with homogenous glycosylation and improved effector functions. Central to this strategy is the use of enzymes with activity on the Asn297 glycan. EndoS and EndoS2, produced by Streptococcus pyogenes, are endoglycosidases with remarkable specificity for Asn297 glycosylation, making them ideal tools for chemoenzymatic synthesis. Although both enzymes are specific for IgG, EndoS2 recognizes a wider range of glycans than EndoS. Recent progress has been made in understanding the structural basis for their activities on antibodies. In this review, we examine the molecular mechanism of glycosidic bond cleavage by these enzymes and how specific point mutations convert them into glycosynthases. We also discuss the structural basis for differences in the glycan repertoire that IgG-active endoglycosidases recognize, which focuses on the structure of the loops within the glycoside hydrolase (GH) domain. Finally, we discuss the important contributions of carbohydrate binding modules (CBMs) to endoglycosidase activity, and how CBMs work in concert with GH domains to produce optimal activity on IgG.


Asunto(s)
Glicósido Hidrolasas/metabolismo , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Polisacáridos/metabolismo , Animales , Glicosilación , Humanos , Modelos Moleculares , Estructura Molecular , Polisacáridos/química , Streptococcus pyogenes/enzimología
12.
Artículo en Inglés | MEDLINE | ID: mdl-30642934

RESUMEN

The spread of multidrug or extensively drug-resistant Gram-negative bacteria is a serious public health issue. There are too few new antibiotics in development to combat the threat of multidrug-resistant infections, and consequently the rate of increasing antibiotic resistance is outpacing the drug development process. This fundamentally threatens our ability to treat common infectious diseases. Fosfomycin (FOM) has an established track record of safety in humans and is highly active against Escherichia coli, including multidrug-resistant strains. However, many other Gram-negative pathogens, including the "priority pathogens" Klebsiella pneumoniae and Pseudomonas aeruginosa, are inherently resistant to FOM due to the chromosomal fosA gene, which directs expression of a metal-dependent glutathione S-transferase (FosA) that metabolizes FOM. In this study, we describe the discovery and biochemical and structural characterization of ANY1 (3-bromo-6-[3-(3-bromo-2-oxo-1H-pyrazolo[1,5-a]pyrimidin-6-yl)-4-nitro-1H-pyrazol-5-yl]-1H-pyrazolo[1,5-a]pyrimidin-2-one), a small-molecule active-site inhibitor of FosA. Importantly, ANY1 potentiates FOM activity in representative Gram-negative pathogens. Collectively, our study outlines a new strategy to expand FOM activity to a broader spectrum of Gram-negative pathogens, including multidrug-resistant strains.


Asunto(s)
Antibacterianos/farmacología , Proteínas de Escherichia coli/antagonistas & inhibidores , Escherichia coli/efectos de los fármacos , Fosfomicina/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Farmacorresistencia Bacteriana/fisiología , Farmacorresistencia Bacteriana Múltiple , Humanos , Pruebas de Sensibilidad Microbiana
13.
Am J Physiol Heart Circ Physiol ; 316(6): H1480-H1494, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30978132

RESUMEN

Effector CD8 T cells infiltrate atherosclerotic lesions and are correlated with cardiovascular events, but the mechanisms regulating their recruitment and retention are not well understood. CD137 (4-1BB) is a costimulatory receptor induced on immune cells and expressed at sites of human atherosclerotic plaque. Genetic variants associated with decreased CD137 expression correlate with carotid-intimal thickness and its deficiency in animal models attenuates atherosclerosis. These effects have been attributed in part to endothelial responses to low and disturbed flow (LDF), but CD137 also generates robust effector CD8 T cells as a costimulatory signal. Thus, we asked whether CD8 T cell-specific CD137 stimulation contributes to their infiltration, retention, and IFNγ production in early atherogenesis. We tested this through adoptive transfer of CD8 T cells into recipient C57BL/6J mice that were then antigen primed and CD137 costimulated. We analyzed atherogenic LDF vessels in normolipidemic and PCSK9-mediated hyperlipidemic models and utilized a digestion protocol that allowed for lesional T-cell characterization via flow cytometry and in vitro stimulation. We found that CD137 activation, specifically of effector CD8 T cells, triggers their intimal infiltration into LDF vessels and promotes a persistent innate-like proinflammatory program. Residence of CD137+ effector CD8 T cells further promoted infiltration of endogenous CD8 T cells with IFNγ-producing potential, whereas CD137-deficient CD8 T cells exhibited impaired vessel infiltration, minimal IFNγ production, and reduced infiltration of endogenous CD8 T cells. Our studies thus provide novel insight into how CD137 costimulation of effector T cells, independent of plaque-antigen recognition, instigates their retention and promotes innate-like responses from immune infiltrates within atherogenic foci. NEW & NOTEWORTHY Our studies identify CD137 costimulation as a stimulus for effector CD8 T-cell infiltration and persistence within atherogenic foci, regardless of atherosclerotic-antigen recognition. These costimulated effector cells, which are generated in pathological states such as viral infection and autoimmunity, have innate-like proinflammatory programs in circulation and within the atherosclerotic microenvironment, providing mechanistic context for clinical correlations of cardiovascular morbidity with increased CD8 T-cell infiltration and markers of activation in the absence of established antigen specificity.


Asunto(s)
Aorta Abdominal/metabolismo , Aterosclerosis/metabolismo , Linfocitos T CD8-positivos/metabolismo , Arterias Carótidas/metabolismo , Inmunidad Innata , Activación de Linfocitos , Placa Aterosclerótica , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Traslado Adoptivo , Animales , Aorta Abdominal/inmunología , Aorta Abdominal/patología , Aterosclerosis/inmunología , Aterosclerosis/patología , Linfocitos T CD8-positivos/inmunología , Arterias Carótidas/inmunología , Arterias Carótidas/patología , Células Cultivadas , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Hiperlipidemias/complicaciones , Interferón gamma/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo , Transducción de Señal , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/deficiencia , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
14.
Adv Exp Med Biol ; 1149: 57-75, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31016628

RESUMEN

Helicobacter pylori adherence to host epithelial cells is essential for its survival against the harsh conditions of the stomach and for successful colonization. Adherence of H. pylori is achieved through several related families of outer membrane proteins and proteins of a type IV secretion system (T4SS), which bridge H. pylori to host cells through protein-protein and other protein-ligand interactions. Local environmental conditions such as cell type, available host cell surface proteins and/or ligands, as well as responses by the host immune system force H. pylori to alter expression of these proteins to adapt quickly to the local environment in order to colonize and survive. Some of these host-pathogen interactions appear to function in a "catch-and-release" manner, regulated by reversible binding at varying pH and allowing H. pylori to detach itself from cells or debris sloughed off the gastric epithelial lining in order to return for subsequent productive interactions. Other interactions between bacterial adhesin proteins and host adhesion molecules, however, appear to function as a committed step in certain pathogenic processes, such as translocation of the CagA oncoprotein through the H. pylori T4SS and into host gastric epithelial cells. Understanding these adhesion interactions is critical for devising new therapeutic strategies, as they are responsible for the earliest stage of infection and its maintenance. This review will discuss the expression and regulation of several outer membrane proteins and CagL, how they engage their known host cell protein/ligand targets, and their effects on clinical outcome.


Asunto(s)
Adhesión Bacteriana , Células Epiteliales , Infecciones por Helicobacter , Helicobacter pylori , Interacciones Huésped-Patógeno , Estómago , Adhesinas Bacterianas/metabolismo , Células Epiteliales/microbiología , Humanos , Estómago/microbiología
16.
J Virol ; 91(20)2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28794021

RESUMEN

Despite recent advances in therapeutic options, hepatitis C virus (HCV) remains a severe global disease burden, and a vaccine can substantially reduce its incidence. Due to its extremely high sequence variability, HCV can readily escape the immune response; thus, an effective vaccine must target conserved, functionally important epitopes. Using the structure of a broadly neutralizing antibody in complex with a conserved linear epitope from the HCV E2 envelope glycoprotein (residues 412 to 423; epitope I), we performed structure-based design of immunogens to induce antibody responses to this epitope. This resulted in epitope-based immunogens based on a cyclic defensin protein, as well as a bivalent immunogen with two copies of the epitope on the E2 surface. We solved the X-ray structure of a cyclic immunogen in complex with the HCV1 antibody and confirmed preservation of the epitope conformation and the HCV1 interface. Mice vaccinated with our designed immunogens produced robust antibody responses to epitope I, and their serum could neutralize HCV. Notably, the cyclic designs induced greater epitope-specific responses and neutralization than the native peptide epitope. Beyond successfully designing several novel HCV immunogens, this study demonstrates the principle that neutralizing anti-HCV antibodies can be induced by epitope-based, engineered vaccines and provides the basis for further efforts in structure-based design of HCV vaccines.IMPORTANCE Hepatitis C virus is a leading cause of liver disease and liver cancer, with approximately 3% of the world's population infected. To combat this virus, an effective vaccine would have distinct advantages over current therapeutic options, yet experimental vaccines have not been successful to date, due in part to the virus's high sequence variability leading to immune escape. In this study, we rationally designed several vaccine immunogens based on the structure of a conserved epitope that is the target of broadly neutralizing antibodies. In vivo results in mice indicated that these antigens elicited epitope-specific neutralizing antibodies, with various degrees of potency and breadth. These promising results suggest that a rational design approach can be used to generate an effective vaccine for this virus.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Epítopos/inmunología , Hepacivirus/inmunología , Vacunas contra Hepatitis Viral/química , Vacunas contra Hepatitis Viral/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Diseño de Fármacos , Epítopos/química , Ratones , Proteínas del Envoltorio Viral/inmunología , Vacunas contra Hepatitis Viral/administración & dosificación
17.
Biochem J ; 474(4): 517-519, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28159897

RESUMEN

Neisseria meningitidis is a Gram-negative bacterium capable of causing deadly invasive disease. Two recently developed vaccines against N. meningitidis serogroup B include recombinant factor H binding protein (fHbp), a surface protein that meningococci use to evade the host immune system. Many anti-fHbp monoclonal antibodies (mAbs) produced against fHbp fail to trigger complement-mediated bacteriolysis when used alone in vitro, but are highly synergistic and bactericidal when used in combination. This opened the door to defining the structural basis by which mAbs activate complement synergistically when binding to different epitopes on the same antigen, a story that is told by Malito et al. in a recent issue of the Biochemical Journal. Using two separate crystal structures of fHbp bound to Fabs from synergistic mAbs, they were able to model the structure of both full length antibodies bound simultaneously to fHbp. This revealed that the bound antibodies orient their Fc domains 115-130 Å apart, a distance that is compatible with multivalent C1q binding. The need for a precise orientation of Fc domains in order to efficiently activate effector functions is an emerging theme across multiple fields, and its implications could have broad impacts on vaccinology and immunotherapy.


Asunto(s)
Anticuerpos Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Antígenos Bacterianos/química , Proteínas Bacterianas/química , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/química , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/química , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/química , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/inmunología , Sitios de Unión de Anticuerpos , Complemento C1q/química , Complemento C1q/metabolismo , Cristalografía por Rayos X , Sinergismo Farmacológico , Quimioterapia Combinada , Epítopos/química , Epítopos/inmunología , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Neisseria meningitidis/química , Neisseria meningitidis/efectos de los fármacos , Neisseria meningitidis/inmunología , Unión Proteica
18.
Proc Natl Acad Sci U S A ; 112(44): 13561-6, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26483485

RESUMEN

Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) comprise a large family of cell surface adhesion molecules that bind to themselves and other family members to carry out numerous cellular functions, including proliferation, signaling, differentiation, tumor suppression, and survival. They also play diverse and significant roles in immunity and infection. The formation of CEACAM oligomers is caused predominantly by interactions between their N-terminal IgV domains. Although X-ray crystal structures of CEACAM IgV domain homodimers have been described, how CEACAMs form heterodimers or remain monomers is poorly understood. To address this key aspect of CEACAM function, we determined the crystal structures of IgV domains that form a homodimeric CEACAM6 complex, monomeric CEACAM8, and a heterodimeric CEACAM6-CEACAM8 complex. To confirm and quantify these interactions in solution, we used analytical ultracentrifugation to measure the dimerization constants of CEACAM homodimers and isothermal titration calorimetry to determine the thermodynamic parameters and binding affinities of CEACAM heterodimers. We found the CEACAM6-CEACAM8 heterodimeric state to be substantially favored energetically relative to the CEACAM6 homodimer. Our data provide a molecular basis for the adoption of the diverse oligomeric states known to exist for CEACAMs and suggest ways in which CEACAM6 and CEACAM8 regulate the biological functions of one another, as well as of additional CEACAMs with which they interact, both in cis and in trans.


Asunto(s)
Antígenos CD/química , Moléculas de Adhesión Celular/química , Multimerización de Proteína , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Antígenos CD/genética , Antígenos CD/metabolismo , Calorimetría/métodos , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Cristalografía por Rayos X , Células Epiteliales/metabolismo , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Granulocitos/metabolismo , Células HEK293 , Humanos , Enlace de Hidrógeno , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Homología de Secuencia de Aminoácido
19.
J Biol Chem ; 291(44): 22924-22935, 2016 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-27634041

RESUMEN

Acinetobacter baumannii is a Gram-negative coccobacillus found primarily in hospital settings that has recently emerged as a source of hospital-acquired infections. A. baumannii expresses a variety of virulence factors, including type IV pili, bacterial extracellular appendages often essential for attachment to host cells. Here, we report the high resolution structures of the major pilin subunit, PilA, from three Acinetobacter strains, demonstrating that A. baumannii subsets produce morphologically distinct type IV pilin glycoproteins. We examine the consequences of this heterogeneity for protein folding and assembly as well as host-cell adhesion by Acinetobacter Comparisons of genomic and structural data with pilin proteins from other species of soil gammaproteobacteria suggest that these structural differences stem from evolutionary pressure that has resulted in three distinct classes of type IVa pilins, each found in multiple species.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Proteínas Bacterianas/química , Fimbrias Bacterianas/química , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/química , Acinetobacter baumannii/clasificación , Acinetobacter baumannii/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana Múltiple , Evolución Molecular , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Gammaproteobacteria/química , Gammaproteobacteria/clasificación , Gammaproteobacteria/aislamiento & purificación , Gammaproteobacteria/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Modelos Moleculares , Filogenia , Microbiología del Suelo
20.
Artículo en Inglés | MEDLINE | ID: mdl-28874374

RESUMEN

Fosfomycin exhibits broad-spectrum antibacterial activity and is being reevaluated for the treatment of extensively drug-resistant pathogens. Its activity in Gram-negative organisms, however, can be compromised by expression of FosA, a metal-dependent transferase that catalyzes the conjugation of glutathione to fosfomycin, rendering the antibiotic inactive. In this study, we solved the crystal structures of two of the most clinically relevant FosA enzymes: plasmid-encoded FosA3 from Escherichia coli and chromosomally encoded FosA from Klebsiella pneumoniae (FosAKP). The structure, molecular dynamics, catalytic activity, and fosfomycin resistance of FosA3 and FosAKP were also compared to those of FosA from Pseudomonas aeruginosa (FosAPA), for which prior crystal structures exist. E. coli TOP10 transformants expressing FosA3 and FosAKP conferred significantly greater fosfomycin resistance (MIC, >1,024 µg/ml) than those expressing FosAPA (MIC, 16 µg/ml), which could be explained in part by the higher catalytic efficiencies of the FosA3 and FosAKP enzymes. Interestingly, these differences in enzyme activity could not be attributed to structural differences at their active sites. Instead, molecular dynamics simulations and hydrogen-deuterium exchange experiments with FosAKP revealed dynamic interconnectivity between its active sites and a loop structure that extends from the active site of each monomer and traverses the dimer interface. This dimer interface loop is longer and more extended in FosAKP and FosA3 than in FosAPA, and kinetic analyses of FosAKP and FosAPA loop-swapped chimeric enzymes highlighted its importance in FosA activity. Collectively, these data yield novel insights into fosfomycin resistance that could be leveraged to develop new strategies to inhibit FosA and potentiate fosfomycin activity.


Asunto(s)
Farmacorresistencia Bacteriana/fisiología , Proteínas de Escherichia coli/química , Escherichia coli/efectos de los fármacos , Fosfomicina/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Dominio Catalítico , Cristalografía por Rayos X , Medición de Intercambio de Deuterio , Farmacorresistencia Bacteriana/efectos de los fármacos , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Klebsiella pneumoniae/genética , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Potasio/metabolismo , Multimerización de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA