Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(16): e89, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37548398

RESUMEN

We describe a novel method for in vitro protein display-click display-that does not depend on maintaining RNA integrity during biopanning and yields covalently linked protein-cDNA complexes from double-stranded input DNA within 2 h. The display is achieved in a one-pot format encompassing transcription, translation and reverse transcription reactions in series. Stable linkage between proteins and the encoding cDNA is mediated by a modified DNA linker-ML-generated via a click chemistry reaction between a puromycin-containing oligo and a cDNA synthesis primer. Biopanning of a click-displayed mock library coupled with next-generation sequencing analysis revealed >600-fold enrichment of target binders within a single round of panning. A synthetic library of Designed Ankyrin Repeat Proteins (DARPins) with ∼1012 individual members was generated using click display in a 25-µl reaction and six rounds of library panning against a model protein yielded a panel of nanomolar binders. This study establishes click display as a powerful tool for protein binder discovery/engineering and provides a convenient platform for in vitro biopanning selection even in RNase-rich environments such as on whole cells.


Asunto(s)
Evolución Molecular Dirigida , Biblioteca de Péptidos , ADN/química , ADN Complementario/genética , Ingeniería de Proteínas , Proteínas/genética , Evolución Molecular Dirigida/métodos
2.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443167

RESUMEN

The blood-clotting protein fibrinogen has been implicated in host defense following Staphylococcus aureus infection, but precise mechanisms of host protection and pathogen clearance remain undefined. Peritonitis caused by staphylococci species is a complication for patients with cirrhosis, indwelling catheters, or undergoing peritoneal dialysis. Here, we sought to characterize possible mechanisms of fibrin(ogen)-mediated antimicrobial responses. Wild-type (WT) (Fib+) mice rapidly cleared S. aureus following intraperitoneal infection with elimination of ∼99% of an initial inoculum within 15 min. In contrast, fibrinogen-deficient (Fib-) mice failed to clear the microbe. The genotype-dependent disparity in early clearance resulted in a significant difference in host mortality whereby Fib+ mice uniformly survived whereas Fib- mice exhibited high mortality rates within 24 h. Fibrin(ogen)-mediated bacterial clearance was dependent on (pro)thrombin procoagulant function, supporting a suspected role for fibrin polymerization in this mechanism. Unexpectedly, the primary host initiator of coagulation, tissue factor, was found to be dispensable for this antimicrobial activity. Rather, the bacteria-derived prothrombin activator vWbp was identified as the source of the thrombin-generating potential underlying fibrin(ogen)-dependent bacterial clearance. Mice failed to eliminate S. aureus deficient in vWbp, but clearance of these same microbes in WT mice was restored if active thrombin was administered to the peritoneal cavity. These studies establish that the thrombin/fibrinogen axis is fundamental to host antimicrobial defense, offer a possible explanation for the clinical observation that coagulase-negative staphylococci are a highly prominent infectious agent in peritonitis, and suggest caution against anticoagulants in individuals susceptible to peritoneal infections.


Asunto(s)
Fibrinógeno/metabolismo , Peritonitis/metabolismo , Protrombina/metabolismo , Animales , Antibacterianos/metabolismo , Antiinfecciosos/metabolismo , Coagulación Sanguínea , Coagulasa/metabolismo , Femenino , Fibrina/metabolismo , Fibrinógeno/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Tromboplastina
3.
PLoS Pathog ; 17(1): e1009182, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33406160

RESUMEN

Streptococcus gallolyticus subspecies gallolyticus (Sgg) has a strong clinical association with colorectal cancer (CRC) and actively promotes the development of colon tumors. However, the molecular determinants involved in Sgg pathogenicity in the gut are unknown. Bacterial type VII secretion systems (T7SS) mediate pathogen interactions with their host and are important for virulence in pathogenic mycobacteria and Staphylococcus aureus. Through genome analysis, we identified a locus in Sgg strain TX20005 that encodes a putative type VII secretion system (designated as SggT7SST05). We showed that core genes within the SggT7SST05 locus are expressed in vitro and in the colon of mice. Western blot analysis showed that SggEsxA, a protein predicted to be a T7SS secretion substrate, is detected in the bacterial culture supernatant, indicating that this SggT7SST05 is functional. Deletion of SggT7SST05 (TX20005Δesx) resulted in impaired bacterial adherence to HT29 cells and abolished the ability of Sgg to stimulate HT29 cell proliferation. Analysis of bacterial culture supernatants suggest that SggT7SST05-secreted factors are responsible for the pro-proliferative activity of Sgg, whereas Sgg adherence to host cells requires both SggT7SST05-secreted and bacterial surface-associated factors. In a murine gut colonization model, TX20005Δesx showed significantly reduced colonization compared to the parent strain. Furthermore, in a mouse model of CRC, mice exposed to TX20005 had a significantly higher tumor burden compared to saline-treated mice, whereas those exposed to TX20005Δesx did not. Examination of the Sgg load in the colon in the CRC model suggests that SggT7SST05-mediated activities are directly involved in the promotion of colon tumors. Taken together, these results reveal SggT7SST05 as a previously unrecognized pathogenicity determinant for Sgg colonization of the colon and promotion of colon tumors.


Asunto(s)
Proliferación Celular , Neoplasias del Colon/patología , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus gallolyticus subspecies gallolyticus/fisiología , Sistemas de Secreción Tipo VII/metabolismo , Animales , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/microbiología , Humanos , Ratones , Ratones Endogámicos A , Infecciones Estreptocócicas/metabolismo
4.
Mol Microbiol ; 115(6): 1207-1228, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33325565

RESUMEN

Catabolite control protein A (CcpA) is a master regulator of carbon source utilization and contributes to the virulence of numerous medically important Gram-positive bacteria. Most functional assessments of CcpA, including interaction with its key co-factor HPr, have been performed in nonpathogenic bacteria. In this study we aimed to identify the in vivo DNA binding profile of CcpA and assess the extent to which HPr is required for CcpA-mediated regulation and DNA binding in the major human pathogen group A Streptococcus (GAS). Using a combination RNAseq/ChIP-seq approach, we found that CcpA affects transcript levels of 514 of 1667 GAS genes (31%) whereas direct DNA binding was identified for 105 GAS genes. Three of the directly regulated genes encode the key GAS virulence factors Streptolysin S, PrtS (IL-8 degrading proteinase), and SpeB (cysteine protease). Mutating CcpA Val301 to Ala (strain 2221-CcpA-V301A) abolished interaction between CcpA and HPr and impacted the transcript levels of 205 genes (40%) in the total CcpA regulon. By ChIP-seq analysis, CcpAV301A bound to DNA from 74% of genes bound by wild-type CcpA, but generally with lower affinity. These data delineate the direct CcpA regulon and clarify the HPr-dependent and independent activities of CcpA in a key pathogenic bacterium.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Streptococcus pyogenes/metabolismo , Proteínas Bacterianas/genética , Proteínas Portadoras/metabolismo , Cromatina/genética , ADN Bacteriano/genética , Proteínas de Unión al ADN/genética , Exotoxinas/genética , Genoma Bacteriano/genética , Unión Proteica/fisiología , RNA-Seq , Proteínas Represoras/metabolismo , Serina Endopeptidasas/genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Estreptolisinas/genética , Virulencia/genética , Factores de Virulencia/genética
5.
PLoS Pathog ; 15(6): e1007816, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31216354

RESUMEN

Fibrinogen is an essential part of the blood coagulation cascade and a major component of the extracellular matrix in mammals. The interface between fibrinogen and bacterial pathogens is an important determinant of the outcome of infection. Here, we demonstrate that a canine host-restricted skin pathogen, Staphylococcus pseudintermedius, produces a cell wall-associated protein (SpsL) that has evolved the capacity for high strength binding to canine fibrinogen, with reduced binding to fibrinogen of other mammalian species including humans. Binding occurs via the surface-expressed N2N3 subdomains, of the SpsL A-domain, to multiple sites in the fibrinogen α-chain C-domain by a mechanism analogous to the classical dock, lock, and latch binding model. Host-specific binding is dependent on a tandem repeat region of the fibrinogen α-chain, a region highly divergent between mammals. Of note, we discovered that the tandem repeat region is also polymorphic in different canine breeds suggesting a potential influence on canine host susceptibility to S. pseudintermedius infection. Importantly, the strong host-specific fibrinogen-binding interaction of SpsL to canine fibrinogen is essential for bacterial aggregation and biofilm formation, and promotes resistance to neutrophil phagocytosis, suggesting a key role for the interaction during pathogenesis. Taken together, we have dissected a bacterial surface protein-ligand interaction resulting from the co-evolution of host and pathogen that promotes host-specific innate immune evasion and may contribute to its host-restricted ecology.


Asunto(s)
Proteínas Bacterianas/inmunología , Biopelículas/crecimiento & desarrollo , Fibrinógeno/inmunología , Evasión Inmune , Inmunidad Innata , Staphylococcus/fisiología , Animales , Proteínas Bacterianas/genética , Pollos , Perros , Fibrinógeno/genética , Humanos
6.
J Biol Chem ; 292(21): 8797-8810, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28400484

RESUMEN

The Staphylococcus aureus cell surface contains cell wall-anchored proteins such as fibronectin-binding protein A (FnBPA) that bind to host ligands (e.g. fibronectin; Fn) present in the extracellular matrix of tissue or coatings on cardiac implants. Recent clinical studies have found a correlation between cardiovascular infections caused by S. aureus and nonsynonymous SNPs in FnBPA. Atomic force microscopy (AFM), surface plasmon resonance (SPR), and molecular simulations were used to investigate interactions between Fn and each of eight 20-mer peptide variants containing amino acids Ala, Asn, Gln, His, Ile, and Lys at positions equivalent to 782 and/or 786 in Fn-binding repeat-9 of FnBPA. Experimentally measured bond lifetimes (1/koff) and dissociation constants (Kd = koff/kon), determined by mechanically dissociating the Fn·peptide complex at loading rates relevant to the cardiovascular system, varied from the lowest-affinity H782A/K786A peptide (0.011 s, 747 µm) to the highest-affinity H782Q/K786N peptide (0.192 s, 15.7 µm). These atomic force microscopy results tracked remarkably well to metadynamics simulations in which peptide detachment was defined solely by the free-energy landscape. Simulations and SPR experiments suggested that an Fn conformational change may enhance the stability of the binding complex for peptides with K786I or H782Q/K786I (Kdapp = 0.2-0.5 µm, as determined by SPR) compared with the lowest-affinity double-alanine peptide (Kdapp = 3.8 µm). Together, these findings demonstrate that amino acid substitutions in Fn-binding repeat-9 can significantly affect bond strength and influence the conformation of Fn upon binding. They provide a mechanistic explanation for the observation of nonsynonymous SNPs in fnbA among clinical isolates of S. aureus that cause endovascular infections.


Asunto(s)
Adhesinas Bacterianas/química , Adhesinas Bacterianas/genética , Polimorfismo de Nucleótido Simple , Staphylococcus aureus/química , Staphylococcus aureus/genética , Adhesinas Bacterianas/metabolismo , Sustitución de Aminoácidos , Microscopía de Fuerza Atómica , Mutación Missense , Secuencias Repetitivas de Aminoácido , Staphylococcus aureus/metabolismo , Resonancia por Plasmón de Superficie
7.
PLoS Pathog ; 12(1): e1005404, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26808924

RESUMEN

Pathogens that traffic in blood, lymphatics, or interstitial fluids must adopt strategies to evade innate immune defenses, notably the complement system. Through recruitment of host regulators of complement to their surface, many pathogens are able to escape complement-mediated attack. The Lyme disease spirochete, Borrelia burgdorferi, produces a number of surface proteins that bind to factor H related molecules, which function as the dominant negative regulator of the alternative pathway of complement. Relatively less is known about how B. burgdorferi evades the classical pathway of complement despite the observation that some sensu lato strains are sensitive to classical pathway activation. Here we report that the borrelial lipoprotein BBK32 potently and specifically inhibits the classical pathway by binding with high affinity to the initiating C1 complex of complement. In addition, B. burgdorferi cells that produce BBK32 on their surface bind to both C1 and C1r and a serum sensitive derivative of B. burgdorferi is protected from killing via the classical pathway in a BBK32-dependent manner. Subsequent biochemical and biophysical approaches localized the anti-complement activity of BBK32 to its globular C-terminal domain. Mechanistic studies reveal that BBK32 acts by entrapping C1 in its zymogen form by binding and inhibiting the C1 subcomponent, C1r, which serves as the initiating serine protease of the classical pathway. To our knowledge this is the first report of a spirochetal protein acting as a direct inhibitor of the classical pathway and is the only example of a biomolecule capable of specifically and noncovalently inhibiting C1/C1r. By identifying a unique mode of complement evasion this study greatly enhances our understanding of how pathogens subvert and potentially manipulate host innate immune systems.


Asunto(s)
Proteínas Bacterianas/inmunología , Borrelia burgdorferi/inmunología , Activación de Complemento/inmunología , Vía Clásica del Complemento/inmunología , Interacciones Huésped-Parásitos/inmunología , Evasión Inmune/inmunología , Complemento C1/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Inmunoprecipitación , Enfermedad de Lyme/inmunología
8.
Blood ; 126(17): 2047-58, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26228483

RESUMEN

Fibrin(ogen) is central to hemostasis and thrombosis and also contributes to multiple physiologic and pathologic processes beyond coagulation. However, the precise contribution of soluble fibrinogen vs insoluble fibrin matrices to vascular integrity, tissue repair, inflammation, and disease has been undefined and unapproachable. To establish the means to distinguish fibrinogen- and fibrin-dependent processes in vivo, Fib(AEK) mice were generated that carry normal levels of circulating fibrinogen but lack the capacity for fibrin polymer formation due to a germ-line mutation in the Aα chain thrombin cleavage site. Homozygous Fib(AEK) mice developed to term and exhibited postnatal survival superior to that of fibrinogen-deficient mice. Unlike fibrinogen-deficient mice, platelet-rich plasma from Fib(AEK) mice supported normal platelet aggregation in vitro, highlighting that fibrinogen(AEK) retains the functional capacity to support interactions with platelets. Thrombin failed to release fibrinopeptide-A from fibrinogen(AEK) and failed to induce polymer formation with Fib(AEK) plasma or purified fibrinogen(AEK) in 37°C mixtures regardless of incubation time. Fib(AEK) mice displayed both an absence of fibrin polymer formation following liver injury, as assessed by electron microscopy, and a failure to generate stable occlusive thrombi following FeCl3 injury of carotid arteries. Fib(AEK) mice exhibited a profound impediment in Staphylococcus aureus clearance following intraperitoneal infection similar to fibrinogen-deficient mice, yet Fib(AEK) mice displayed a significant infection dose-dependent survival advantage over fibrinogen-deficient mice following peritonitis challenge. Collectively, these findings establish for the first time that fibrin polymer is the molecular form critical for antimicrobial mechanisms while simultaneously highlighting biologically meaningful contributions and functions of the soluble molecule.


Asunto(s)
Fibrina/metabolismo , Fibrinógeno/fisiología , Interacciones Huésped-Patógeno , Mutación/genética , Peritonitis/etiología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Pruebas de Coagulación Sanguínea , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica , Hemostáticos , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Peritonitis/patología , Agregación Plaquetaria , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología
9.
Mol Microbiol ; 96(1): 68-83, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25560615

RESUMEN

Borrelia burgdorferi, the etiologic agent of Lyme disease, adapts to the mammalian hosts by differentially expressing several genes in the BosR and Rrp2-RpoN-RpoS dependent pathways, resulting in a distinct protein profile relative to that seen for survival in the Ixodes spp. tick. Previous studies indicate that a putative lipoprotein, BBA33, is produced in an RpoS-dependent manner under conditions that mimic the mammalian component of the borrelial lifecycle. However, the significance and function for BBA33 is not known. Given its linkage to the BosR/Rrp2-RpoN-RpoS regulatory cascade, we hypothesized that BBA33 facilitates B. burgdorferi infection in the mammalian host. The deletion of bba33 eliminated B. burgdorferi infectivity in C3H mice, which was rescued by genetic complementation with intact bba33. With regard to function, a combinatorial peptide approach, coupled with subsequent in vitro binding assays, indicated that BBA33 binds to collagen type VI and, to a lesser extent, collagen type IV. Whole cell binding assays demonstrated BBA33-dependent binding to human collagen type VI. Taken together, these results suggest that BBA33 interacts with collagenous structures and may function as an adhesin in a process that is required to prevent bacterial clearance.


Asunto(s)
Proteínas Bacterianas/metabolismo , Borrelia burgdorferi/metabolismo , Borrelia burgdorferi/patogenicidad , Colágeno/metabolismo , Regulación Bacteriana de la Expresión Génica , Lipoproteínas/metabolismo , Adhesinas Bacterianas/metabolismo , Animales , Proteínas Bacterianas/genética , Borrelia burgdorferi/genética , Eliminación de Gen , Perfilación de la Expresión Génica , Prueba de Complementación Genética , Humanos , Lipoproteínas/genética , Ratones , Ratones Endogámicos C3H , Unión Proteica
10.
J Bacteriol ; 197(5): 882-92, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25512313

RESUMEN

The WxL domain recently has been identified as a novel cell wall binding domain found in numerous predicted proteins within multiple Gram-positive bacterial species. However, little is known about the function of proteins containing this novel domain. Here, we identify and characterize 6 Enterococcus faecium proteins containing the WxL domain which, by reverse transcription-PCR (RT-PCR) and genomic analyses, are located in three similarly organized operons, deemed WxL loci A, B, and C. Western blotting, electron microscopy, and enzyme-linked immunosorbent assays (ELISAs) determined that genes of WxL loci A and C encode antigenic, cell surface proteins exposed at higher levels in clinical isolates than in commensal isolates. Secondary structural analyses of locus A recombinant WxL domain-containing proteins found they are rich in ß-sheet structure and disordered segments. Using Biacore analyses, we discovered that recombinant WxL proteins from locus A bind human extracellular matrix proteins, specifically type I collagen and fibronectin. Proteins encoded by locus A also were found to bind to each other, suggesting a novel cell surface complex. Furthermore, bile salt survival assays and animal models using a mutant from which all three WxL loci were deleted revealed the involvement of WxL operons in bile salt stress and endocarditis pathogenesis. In summary, these studies extend our understanding of proteins containing the WxL domain and their potential impact on colonization and virulence in E. faecium and possibly other Gram-positive bacterial species.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Enterococcus faecium/metabolismo , Matriz Extracelular/metabolismo , Infecciones por Bacterias Grampositivas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Enterococcus faecium/química , Enterococcus faecium/genética , Enterococcus faecium/patogenicidad , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Datos de Secuencia Molecular , Operón , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Alineación de Secuencia , Virulencia
11.
Infect Immun ; 83(12): 4653-61, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26371130

RESUMEN

The interaction between bacteria and fibronectin is believed to play an important role in the pathogenicity of clinically important Gram-positive cocci. In the present study, we identified a gene encoding a predicted fibronectin-binding protein of Enterococcus faecium (fnm), a homologue of Streptococcus pneumoniae pavA, in the genomes of E. faecium strain TX82 and all other sequenced E. faecium isolates. Full-length recombinant Fnm from strain TX82 bound to immobilized fibronectin in a concentration-dependent manner and also appeared to bind collagen type V and laminin, but not other proteins, such as transferrin, heparin, bovine serum albumin, mucin, or collagen IV. We demonstrated that the N-terminal fragment of Fnm is required for full fibronectin binding, since truncation of this region caused a 2.4-fold decrease (P < 0.05) in the adhesion of E. faecium TX82 to fibronectin. Deletion of fnm resulted in a significant reduction (P < 0.001) in the ability of the mutant, TX6128, to bind fibronectin relative to that of the wild-type strain; in situ reconstitution of fnm in the deletion mutant strain restored adherence. In addition, the Δfnm mutant was highly attenuated relative to TX82 (P ≤ 0.0001) in a mixed-inoculum rat endocarditis model. Taken together, these results demonstrate that Fnm affects the adherence of E. faecium to fibronectin and is important in the pathogenesis of experimental endocarditis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endocarditis Bacteriana/microbiología , Enterococcus faecium/patogenicidad , Genoma Bacteriano , Infecciones por Bacterias Grampositivas/microbiología , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Colágeno Tipo V/metabolismo , Endocarditis Bacteriana/metabolismo , Endocarditis Bacteriana/patología , Enterococcus faecium/genética , Fibronectinas/metabolismo , Infecciones por Bacterias Grampositivas/metabolismo , Infecciones por Bacterias Grampositivas/patología , Proteínas Inmovilizadas/metabolismo , Laminina/metabolismo , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Unión Proteica , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo , Virulencia
12.
Mol Microbiol ; 94(1): 172-85, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25115812

RESUMEN

The molecular pathogenesis of many Staphylococcus aureus infections involves growth of bacteria as biofilm. In addition to polysaccharide intercellular adhesin (PIA) and extracellular DNA, surface proteins appear to mediate the transition of bacteria from planktonic growth to sessile lifestyle as well as biofilm growth, and can enable these processes even in the absence of PIA expression. However, the molecular mechanisms by which surface proteins contribute to biofilm formation are incompletely understood. Here we demonstrate that self-association of the serine-aspartate repeat protein SdrC promotes both bacterial adherence to surfaces and biofilm formation. However, this homophilic interaction is not required for the attachment of bacteria to abiotic surfaces. We identified the subdomain that mediates SdrC dimerization and subsequent cell-cell interactions. In addition, we determined that two adjacently located amino acid sequences within this subdomain are required for the SdrC homophilic interaction. Comparative amino acid sequence analysis indicated that these binding sites are conserved. In summary, our study identifies SdrC as a novel molecular determinant in staphylococcal biofilm formation and describes the mechanism responsible for intercellular interactions. Furthermore, these findings contribute to a growing body of evidence suggesting that homophilic interactions between surface proteins present on neighbouring bacteria induce biofilm growth.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Biopelículas , Staphylococcus aureus/fisiología , Secuencia de Aminoácidos , Adhesión Bacteriana , Proteínas Bacterianas/genética , Dimerización , Humanos , Datos de Secuencia Molecular , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/química , Staphylococcus aureus/genética
13.
PLoS Pathog ; 9(12): e1003816, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24348255

RESUMEN

Upon contact with human plasma, bacteria are rapidly recognized by the complement system that labels their surface for uptake and clearance by phagocytic cells. Staphylococcus aureus secretes the 16 kD Extracellular fibrinogen binding protein (Efb) that binds two different plasma proteins using separate domains: the Efb N-terminus binds to fibrinogen, while the C-terminus binds complement C3. In this study, we show that Efb blocks phagocytosis of S. aureus by human neutrophils. In vitro, we demonstrate that Efb blocks phagocytosis in plasma and in human whole blood. Using a mouse peritonitis model we show that Efb effectively blocks phagocytosis in vivo, either as a purified protein or when produced endogenously by S. aureus. Mutational analysis revealed that Efb requires both its fibrinogen and complement binding residues for phagocytic escape. Using confocal and transmission electron microscopy we show that Efb attracts fibrinogen to the surface of complement-labeled S. aureus generating a 'capsule'-like shield. This thick layer of fibrinogen shields both surface-bound C3b and antibodies from recognition by phagocytic receptors. This information is critical for future vaccination attempts, since opsonizing antibodies may not function in the presence of Efb. Altogether we discover that Efb from S. aureus uniquely escapes phagocytosis by forming a bridge between a complement and coagulation protein.


Asunto(s)
Proteínas Bacterianas/metabolismo , Complemento C3b/metabolismo , Fibrinógeno/metabolismo , Evasión Inmune , Fagocitosis/inmunología , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo , Animales , Factores de Coagulación Sanguínea/metabolismo , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo
14.
Blood ; 121(10): 1783-94, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23299312

RESUMEN

Fibrinogen can support host antimicrobial containment/clearance mechanisms, yet selected pathogens appear to benefit from host procoagulants to drive bacterial virulence. Here, we explored the hypothesis that host fibrin(ogen), on balance, supports Staphylococcus aureus infection in the context of septicemia. Survival studies following intravenous infection in control and fibrinogen-deficient mice established the overall utility of host fibrin(ogen) to S. aureus virulence. Complementary studies in mice expressing mutant forms of fibrinogen-retaining clotting function, but lacking either the bacterial ClfA (Fibγ(Δ5)) binding motif or the host leukocyte integrin receptor αMß2 (Fibγ(390-396A)) binding motif, revealed the preeminent importance of the bacterial ClfA-fibrin(ogen) interaction in determining host survival. Studies of mice lacking platelets or the platelet integrin receptor subunit αIIb established that the survival benefits observed in Fibγ(Δ5) mice were largely independent of platelet αIIbß3-mediated engagement of fibrinogen. Fibγ(Δ5) mice exhibited reduced bacterial burdens in the hearts and kidneys, a blunted host proinflammatory cytokine response, diminished microscopic tissue damage, and significantly diminished plasma markers of cardiac and other organ damage. These findings indicate that host fibrin(ogen) and bacterial ClfA are dual determinants of virulence and that therapeutic interventions at the level of fibrinogen could be advantageous in S. aureus septicemia.


Asunto(s)
Coagulasa/metabolismo , Fibrinógeno/fisiología , Antígeno de Macrófago-1/fisiología , Sepsis/mortalidad , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Virulencia/fisiología , Afibrinogenemia/etiología , Animales , Adhesión Bacteriana/fisiología , Sitios de Unión , Citocinas/sangre , Citometría de Flujo , Ratones , Ratones Noqueados , Sepsis/etiología , Sepsis/prevención & control , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/metabolismo , Tasa de Supervivencia
15.
J Biol Chem ; 288(28): 20520-31, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23720782

RESUMEN

Members of a family of collagen-binding microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) from Gram-positive bacteria are established virulence factors in several infectious diseases models. Here, we report that these adhesins also can bind C1q and act as inhibitors of the classical complement pathway. Molecular analyses of Cna from Staphylococcus aureus suggested that this prototype MSCRAMM bound to the collagenous domain of C1q and interfered with the interactions of C1r with C1q. As a result, C1r2C1s2 was displaced from C1q, and the C1 complex was deactivated. This novel function of the Cna-like MSCRAMMs represents a potential immune evasion strategy that could be used by numerous Gram-positive pathogens.


Asunto(s)
Adhesinas Bacterianas/inmunología , Activación de Complemento/inmunología , Vía Clásica del Complemento/inmunología , Bacterias Grampositivas/inmunología , Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Unión Competitiva/inmunología , Colágeno/inmunología , Colágeno/metabolismo , Complemento C1q/inmunología , Complemento C1q/metabolismo , Complemento C1r/inmunología , Complemento C1r/metabolismo , Ensayo de Inmunoadsorción Enzimática , Bacterias Grampositivas/metabolismo , Historia del Siglo XVIII , Humanos , Immunoblotting , Cinética , Modelos Moleculares , Unión Proteica/inmunología , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie
16.
Cell Rep Med ; 5(5): 101530, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38688275

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy is hindered in solid tumor treatment due to the immunosuppressive tumor microenvironment and suboptimal T cell persistence. Current strategies do not address nutrient competition in the microenvironment. Hence, we present a metabolic refueling approach using inosine as an alternative fuel. CAR T cells were engineered to express membrane-bound CD26 and cytoplasmic adenosine deaminase 1 (ADA1), converting adenosine to inosine. Autocrine secretion of ADA1 upon CD3/CD26 stimulation activates CAR T cells, improving migration and resistance to transforming growth factor ß1 suppression. Fusion of ADA1 with anti-CD3 scFv further boosts inosine production and minimizes tumor cell feeding. In mouse models of hepatocellular carcinoma and non-small cell lung cancer, metabolically refueled CAR T cells exhibit superior tumor reduction compared to unmodified CAR T cells. Overall, our study highlights the potential of selective inosine refueling to enhance CAR T therapy efficacy against solid tumors.


Asunto(s)
Adenosina Desaminasa , Dipeptidil Peptidasa 4 , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Animales , Adenosina Desaminasa/metabolismo , Humanos , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Ratones , Inmunoterapia Adoptiva/métodos , Dipeptidil Peptidasa 4/metabolismo , Dipeptidil Peptidasa 4/inmunología , Línea Celular Tumoral , Linfocitos T/inmunología , Linfocitos T/metabolismo , Inosina , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/patología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/patología
17.
J Biol Chem ; 287(41): 34856-65, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22865854

RESUMEN

The collagen-binding bacterial proteins, Ace and Cna, are well characterized on the biochemical and structural level. Despite overall structural similarity, recombinant forms of the Ace and Cna ligand-binding domains exhibit significantly different affinities and binding kinetics for collagen type I (CI) in vitro. In this study, we sought to understand, in submolecular detail, the bases for these differences. Using a structure-based approach, we engineered Cna and Ace variants by altering specific structural elements within the ligand-binding domains. Surface plasmon resonance-based binding analysis demonstrated that mutations that are predicted to alter the orientation of the Ace and Cna N(1) and N(2) subdomains significantly affect the interaction between the MSCRAMM (microbial surface components recognizing adhesive matrix molecule) and CI in vitro, including affinity, association/dissociation rates and binding ratio. Moreover, we utilized this information to engineer an Ace variant with an 11,000-fold higher CI affinity than the parent protein. Finally, we noted that several engineered proteins that exhibited a weak interaction with CI recognized more sites on CI, suggesting an inverse correlation between affinity and specificity.


Asunto(s)
Adhesinas Bacterianas/química , Proteínas Bacterianas/química , Proteínas Portadoras/química , Colágeno/química , Enterococcus faecalis/química , Ingeniería de Proteínas , Staphylococcus aureus/química , Adhesinas Bacterianas/genética , Secuencias de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Colágeno/genética , Enterococcus faecalis/genética , Unión Proteica , Estructura Terciaria de Proteína , Staphylococcus aureus/genética , Relación Estructura-Actividad
18.
Biomacromolecules ; 14(7): 2225-33, 2013 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-23758437

RESUMEN

The highly tunable properties of poly(ethylene glycol) (PEG)-based hydrogel systems permit their use in a wide array of regenerative medicine and drug delivery applications. One of the most valuable properties of PEG hydrogels is their intrinsic resistance to protein adsorption and cell adhesion, as it allows for a controlled introduction of desired bioactive factors including proteins, peptides, and drugs. Acrylate-PEG-N-hydroxysuccinimide (Acr-PEG-NHS) is widely utilized as a PEG linker to functionalize bioactive factors with photo-cross-linkable groups. This enables their facile incorporation into PEG hydrogel networks or the use of PEGylation strategies for drug delivery. However, PEG linkers can sterically block integrin binding sites on functionalized proteins and reduce cell-material interactions. In this study we demonstrate that reducing the density of PEG linkers on protein backbones during functionalization results in significantly improved cell adhesion and spreading to bioactive hydrogels. However, this reduction in functionalization density also increases protein loss from the matrix over time due to ester hydrolysis of the Acr-PEG-NHS linkers. To address this, a novel PEG linker, acrylamide-PEG-isocyanate (Aam-PEG-I), with enhanced hydrolytic stability was synthesized. It was found that decreasing functionalization density with Aam-PEG-I resulted in comparable increases in cell adhesion and spreading to Acr-PEG-NHS systems while maintaining protein and bioactivity levels within the hydrogel network over a significantly longer time frame. Thus, Aam-PEG-I provides a new option for protein functionalization for use in a wide range of applications that improves initial and sustained cell-material interactions to enhance control of bioactivity.


Asunto(s)
Materiales Biocompatibles/química , Adhesión Celular/fisiología , Células Endoteliales/fisiología , Hidrogeles/química , Polietilenglicoles/química , Animales , Aorta/citología , Aorta/fisiología , Bovinos , Comunicación Celular , Células Cultivadas , Sistemas de Liberación de Medicamentos , Células Endoteliales/citología , Regeneración
19.
Front Immunol ; 14: 1221108, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37828992

RESUMEN

Staphylococcus aureus pathology is caused by a plethora of virulence factors able to combat multiple host defence mechanisms. Fibrinogen (Fg), a critical component in the host coagulation cascade, plays an important role in the pathogenesis of this bacterium, as it is the target of numerous staphylococcal virulence proteins. Amongst its secreted virulence factors, coagulase (Coa) and Extracellular fibrinogen-binding protein (Efb) share common Fg binding motives and have been described to form a Fg shield around staphylococcal cells, thereby allowing efficient bacterial spreading, phagocytosis escape and evasion of host immune system responses. Targeting these proteins with monoclonal antibodies thus represents a new therapeutic option against S. aureus. To this end, here we report the selection and characterization of fully human, sequence-defined, monoclonal antibodies selected against the C-terminal of coagulase. Given the functional homology between Coa and Efb, we also investigated if the generated antibodies bound the two virulence factors. Thirteen unique antibodies were isolated from naïve antibodies gene libraries by antibody phage display. As anticipated, most of the selected antibodies showed cross-recognition of these two proteins and among them, four were able to block the interaction between Coa/Efb and Fg. Furthermore, our monoclonal antibodies could interact with the two main Fg binding repeats present at the C-terminal of Coa and distinguish them, suggesting the presence of two functionally different Fg-binding epitopes.


Asunto(s)
Coagulasa , Infecciones Estafilocócicas , Humanos , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Proteínas Bacterianas , Coagulasa/inmunología , Fibrinógeno/química , Fibrinógeno/metabolismo , Fagocitosis , Staphylococcus aureus , Factores de Virulencia/metabolismo , Sitios de Unión de Anticuerpos
20.
Nat Struct Mol Biol ; 30(12): 1878-1892, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37932451

RESUMEN

Emerging evidence suggests that cryptic translation beyond the annotated translatome produces proteins with developmental or physiological functions. However, functions of cryptic non-canonical open reading frames (ORFs) in cancer remain largely unknown. To fill this gap and systematically identify colorectal cancer (CRC) dependency on non-canonical ORFs, we apply an integrative multiomic strategy, combining ribosome profiling and a CRISPR-Cas9 knockout screen with large-scale analysis of molecular and clinical data. Many such ORFs are upregulated in CRC compared to normal tissues and are associated with clinically relevant molecular subtypes. We confirm the in vivo tumor-promoting function of the microprotein SMIMP, encoded by a primate-specific, long noncoding RNA, the expression of which is associated with poor prognosis in CRC, is low in normal tissues and is specifically elevated in CRC and several other cancer types. Mechanistically, SMIMP interacts with the ATPase-forming domains of SMC1A, the core subunit of the cohesin complex, and facilitates SMC1A binding to cis-regulatory elements to promote epigenetic repression of the tumor-suppressive cell cycle regulators encoded by CDKN1A and CDKN2B. Thus, our study reveals a cryptic microprotein as an important component of cohesin-mediated gene regulation and suggests that the 'dark' proteome, encoded by cryptic non-canonical ORFs, may contain potential therapeutic or diagnostic targets.


Asunto(s)
Sistemas CRISPR-Cas , Neoplasias , Animales , Humanos , Sistemas de Lectura Abierta/genética , Sistemas CRISPR-Cas/genética , Neoplasias/genética , Proteoma/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA