Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 231
Filtrar
1.
J Med Chem ; 67(5): 3668-3678, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38308631

RESUMEN

FmlH, a bacterial adhesin of uropathogenic Escherichia coli (UPEC), has been shown to provide a fitness advantage in colonizing the bladder during chronic urinary tract infections (UTIs). Previously reported ortho-biphenyl glycosides based on ßGal and ßGalNAc have excellent binding affinity to FmlH and potently block binding to its natural carbohydrate receptor, but they lack oral bioavailability. In this paper, we outline studies where we have optimized compounds for improved pharmacokinetics, leading to the discovery of novel analogues with good oral bioavailability. We synthesized galactosides with the anomeric O-linker replaced with more stable S- and C-linked linkers. We also investigated modifications to the GalNAc sugar and modifications to the biphenyl aglycone. We identified GalNAc 69 with an IC50 of 0.19 µM against FmlH and 53% oral bioavailability in mice. We also obtained a FimlH-bound X-ray structure of lead compound 69 (AM4085) which has potential as a new antivirulence therapeutic for UTIs.


Asunto(s)
Infecciones por Escherichia coli , Infecciones Urinarias , Escherichia coli Uropatógena , Ratones , Animales , Lectinas , Adhesinas de Escherichia coli/química , Infecciones Urinarias/tratamiento farmacológico , Compuestos de Bifenilo/química , Escherichia coli Uropatógena/metabolismo , Infecciones por Escherichia coli/tratamiento farmacológico
2.
Biochem Biophys Res Commun ; 696: 149534, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38241810

RESUMEN

Autotransporters constitute a large family of natural proteins that are essential for delivering many types of proteins and peptides across the outer membrane in Gram-negative bacteria. In biotechnology, autotransporters have been explored for display of recombinant proteins and peptides on the surface of Escherichia coli and have potential as tools for directed evolution of affinity proteins. Here, we investigate conditions for AIDA-I autotransporter-mediated display of recombinant proteins. A new expression vector was designed and engineered for this purpose, and a panel of proteins from different affinity-protein classes were subcloned to the vector, followed by evaluation of expression, surface display and functionality. Surface expression was explored in ten different E. coli strains together with assessment of transformation efficiencies. Furthermore, the most promising strain and expression vector combination was used in mock library selections for evaluation of magnetic-assisted cell sortings (MACS). The results demonstrated dramatically different performances depending on the type of affinity protein and choice of E. coli strain. The optimized MACS protocol showed efficient enrichment, and thus potential for the new AIDA-I display system to be used in methods for directed evolution of affinity proteins.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Escherichia coli/genética , Escherichia coli/metabolismo , Adhesinas de Escherichia coli/química , Sistemas de Secreción Tipo V/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Péptidos/metabolismo
3.
Proteins ; 92(1): 117-133, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37700555

RESUMEN

The bacterial adhesin FimH is a model for the study of protein allostery because its structure has been resolved in multiple configurations, including the active and the inactive state. FimH consists of a pilin domain (PD) that anchors it to the rest of the fimbria and an allosterically regulated lectin domain (LD) that binds mannose on the surface of infected cells. Under normal conditions, the two domains are docked to each other and LD binds mannose weakly. However, in the presence of tensile force generated by shear the domains separate and conformational changes propagate across LD resulting in a stronger bond to mannose. Recently, the crystallographic structure of a variant of FimH has been resolved, called FimH FocH , where PD contains 10 mutations near the inter-domain interface. Although the X-ray structures of FimH and FimH FocH are almost identical, experimental evidence shows that FimH FocH is activated even in the absence of shear. Here, molecular dynamics simulations combined with the Jarzynski equality were used to investigate the discrepancy between the crystallographic structures and the functional assays. The results indicate that the free energy barrier of the unbinding process between LD and PD is drastically reduced in FimH FocH . Rupture of inter-domain hydrogen bonds involving R166 constitutes a rate limiting step of the domain separation process and occurs more readily in FimH FocH than FimH. In conclusion, the mutations in FimH FocH shift the equilibrium toward an equal occupancy of bound and unbound states for LD and PD by reducing a rate limiting step.


Asunto(s)
Manosa , Simulación de Dinámica Molecular , Manosa/química , Regulación Alostérica , Adhesinas de Escherichia coli/química , Escherichia coli/genética , Proteínas Fimbrias/química , Lectinas/metabolismo
4.
BMC Vet Res ; 19(1): 187, 2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37789311

RESUMEN

BACKGROUND: Avian Escherichia coli (E.coli) type 1 fimbriae adhere to avian tracheal epithelial cells through the FimH protein. However, the adhesion-related antigen is still unknown. The purpose of this study was to analyze the antigenicity of the type 1 fimbrial FimH protein of wild-type avian E. coli, screen antigen epitopes, and prepare monoclonal antibodies (mAbs) that can block the adhesion of avian E. coli. RESULTS: In this study, the nucleic acid homologies of MG2 (O11), TS12 (O18), and YR5 (O78) with K12 were 97.7%, 99.6%, and 97.7%, respectively, and the amino acid sequence similarity reached 98.7%, 99.3%, and 98.0%, respectively. The epitopes and hydrophilicities of the FimH proteins of these three strains were similar. The more obvious lectin domain epitopes were located at FimH protein positions 111-124 and 154-162. The mAbs 7C2 and 7D8 against these two epitopes were prepared. An adhesion inhibition test showed that 7C2 and 7D8 blocked bacterial adhesion to avian tracheal epithelial cells. The mAb 7C2 against the 111-124 epitope inhibited O78 strain adhesion by 93%, and the mAb 7D8 against the 154-162 epitope inhibited O78 strain adhesion by 49%, indicating that these two epitopes are closely related to the adhesion of type 1 fimbriae. However, only the 111-124 epitope-recognizing mAb 7C2 inhibited bacterial agglutination of erythrocytes, indicating that host cell receptor binding and erythrocyte agglutination are not mediated by the same spatial locations within the FimH protein. CONCLUSIONS: The results demonstrate that the mAbs 7C2 and 7D8 against FimH protein positions 111-124 and 154-162 could inhibit the adhesion of E.coli to the chicken trachea.


Asunto(s)
Escherichia coli , Proteínas Fimbrias , Animales , Escherichia coli/genética , Proteínas Fimbrias/genética , Epítopos/metabolismo , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/química , Aglutininas/metabolismo , Adhesión Bacteriana
5.
Ther Innov Regul Sci ; 57(6): 1153-1166, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37578736

RESUMEN

The nature of alpha-D-mannose-natural aldohexose sugar, C-2 glucose epimer, whose intended use is for preventing urinary tract infections-in the interaction with E. coli is addressed in order to drive the issue of its regulatory classification as a medicinal product or medical device. PRISMA systematic review approach was applied; Delphi Panel method was used to target consensus on statements retrieved from evidence. Based on regulatory definitions and research evidence, the mechanism of D-mannose does not involve a metabolic or immunological action while there is uncertainty regarding the pharmacological action. Specific interaction between the product and the bacteria within the body occurs, but its nature is inert: it does not induce a direct response activating or inhibiting body processes. Moreover, the action of D-mannose takes place, even if inside the bladder, outside the epithelium on bacteria that have not yet invaded the urothelial tissue. Therefore, its mechanism of action is not directed to host structures but to structures (bacteria) external to the host's tissues. On the basis of current regulation, the uncertainty as regard a pharmacological action of alpha-D-mannose makes possible its medical device classification: new regulations and legal judgments can add further considerations. From a pharmacological perspective, research is driven versus synthetic mannosides: no further considerations are expected on alpha-D-mannose.


Asunto(s)
Escherichia coli , Manosa , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/metabolismo , Consenso , Escherichia coli/química , Escherichia coli/metabolismo , Proteínas Fimbrias/química , Proteínas Fimbrias/metabolismo , Manosa/química , Manosa/metabolismo , Revisiones Sistemáticas como Asunto
6.
Biophys J ; 122(13): 2744-2756, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37264571

RESUMEN

The bacterial fimbrial adhesin FimH is a remarkable and well-studied catch-bond protein found at the tip of E. coli type 1 pili, which allows pathogenic strains involved in urinary tract infections to bind high-mannose glycans exposed on human epithelia. The catch-bond behavior of FimH, where the strength of the interaction increases when a force is applied to separate the two partners, enables the bacteria to resist clearance when they are subjected to shear forces induced by urine flow. Two decades of experimental studies performed at the single-molecule level, as well as x-ray crystallography and modeling studies, have led to a consensus picture whereby force separates the binding domain from an inhibitor domain, effectively triggering an allosteric conformational change in the former. This force-induced allostery is thought to be responsible for an increased binding affinity at the core of the catch-bond mechanism. However, some important questions remain, the most challenging one being that the crystal structures corresponding to these two allosteric states show almost superimposable binding site geometries, which questions the molecular origin for the large difference in affinity. Using molecular dynamics with a combination of enhanced-sampling techniques, we demonstrate that the static picture provided by the crystal structures conceals a variety of binding site conformations that have a key impact on the apparent affinity. Crucially, the respective populations in each of these conformations are very different between the two allosteric states of the binding domain, which can then be related to experimental affinity measurements. We also evidence a previously unappreciated but important effect: in addition to the well-established role of the force as an allosteric regulator via domain separation, application of force tends to directly favor the high-affinity binding site conformations. We hypothesize that this additional "local" catch-bond effect could delay unbinding between the bacteria and the host cell before the "global" allosteric transition occurs, as well as stabilizing the complex even more once in the high-affinity allosteric state.


Asunto(s)
Escherichia coli , Proteínas Fimbrias , Humanos , Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/metabolismo , Adhesión Bacteriana/fisiología , Sitios de Unión , Unión Proteica
7.
J Biol Chem ; 299(5): 104627, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36944399

RESUMEN

The FimH type-1 fimbrial adhesin allows pathogenic Escherichia coli to adhere to glycoproteins in the epithelial linings of human bladder and intestinal tract, by using multiple fimbriae simultaneously. Pauci- and high-mannose type N-glycans are natural FimH receptors on those glycoproteins. Oligomannose-3 and oligomannose-5 bind with the highest affinity to FimH by using the same Manα1,3Man branch. Oligomannose-6 is generated from oligomannose-5 in the next step of the biogenesis of high-mannose N-glycans, by the transfer of a mannose in α1,2-linkage onto this branch. Using serial crystallography and by measuring the kinetics of binding, we demonstrate that shielding the high-affinity epitope drives the binding of multiple FimH molecules. First, we profiled FimH glycan binding on a microarray containing paucimannosidic N-glycans and in a FimH LEctPROFILE assay. To make the transition to oligomannose-6, we measured the kinetics of FimH binding using paucimannosidic N-glycans, glycoproteins and all four α-dimannosides conjugated to bovine serum albumin. Equimolar mixed interfaces of the dimannosides present in oligomannose-6 and molecular dynamics simulations suggest a positive cooperativity in the bivalent binding of Manα1,3Manα1 and Manα1,6Manα1 dimannosides. The binding of core α1,6-fucosylated oligomannose-3 in cocrystals of FimH is monovalent but interestingly the GlcNAc1-Fuc moiety retains highly flexibility. In cocrystals with oligomannose-6, two FimH bacterial adhesins bind the Manα1,3Manα1 and Manα1,6Manα1 endings of the second trimannose core (A-4'-B). This cooperative switch towards bivalent binding appears sustainable beyond a molar excess of oligomannose-6. Our findings provide important novel structural insights for the design of multivalent FimH antagonists that bind with positive cooperativity.


Asunto(s)
Adhesinas de Escherichia coli , Receptor de Manosa , Modelos Moleculares , Humanos , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/metabolismo , Adhesión Bacteriana , Escherichia coli/metabolismo , Glicoproteínas/metabolismo , Manosa/metabolismo , Receptor de Manosa/química , Receptor de Manosa/metabolismo , Polisacáridos/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Simulación del Acoplamiento Molecular
8.
J Biomol Struct Dyn ; 41(9): 3914-3925, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35403563

RESUMEN

The increase in multidrug-resistant pathogens in urinary tract infections (UTIs) among communities and hospitals threatens our ability to treat these common pathogens. Uropathogenic Escherichia coli (UPEC) strains are the most frequent uropathies linked to the development of UTIs. This work aims to introduce bioactive natural products via virtual screening of small molecules from a public database to prevent biofilm formation by inhibiting FimH, a type 1 fimbriae that plays a crucial role in UPEC pathogenicity. A total of 30926 small molecules from the NPASS database were subjected to screening via molecular docking. Followed by performing in silico ADME studies, seven molecules showed promising docking results ranging from -6.8 to -8.7 kcal/mol. As a result of the docking score findings, 100 ns Molecular dynamics (MD) simulations were performed. Based on MM-PBSA analysis, NPC313334 ligand showed high binding affinity -42 and stability with the binding pocket of FimH protein during molecular dynamic simulations. DFT calculations were also performed on the ligands to calculate the HOMO-LUMO energies of the compounds in order to an idea about their structure and reactivity. This research suggests that NPC313334 may be a possible antibacterial drug candidate that targets FimH to reduce the number of UPEC-related urinary tract infections.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Adhesinas de Escherichia coli , Infecciones Urinarias , Humanos , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/metabolismo , Proteínas Fimbrias/química , Proteínas Fimbrias/metabolismo , Proteínas Fimbrias/uso terapéutico , Simulación del Acoplamiento Molecular , Lectinas , Antibacterianos/farmacología , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología , Infecciones Urinarias/prevención & control
9.
J Mol Biol ; 434(17): 167717, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35798162

RESUMEN

Allosteric proteins transition between 'inactive' and 'active' states. In general, such proteins assume distinct conformational states at the level of secondary, tertiary and/or quaternary structure. Different conformers of an allosteric protein can be antigenically dissimilar and induce antibodies with a highly distinctive specificities and neutralizing functional effects. Here we summarize studies on various functional types of monoclonal antibodies obtained against different allosteric conformers of the mannose-specific bacterial adhesin FimH - the most common cell attachment protein of Escherichia coli and other enterobacterial pathogens. Included are types of antibodies that activate the FimH function via interaction with ligand-induced binding sites or by wedging between domains as well as antibodies that inhibit FimH through orthosteric, parasteric, or novel dynasteric mechanisms. Understanding the molecular mechanism of antibody action against allosteric proteins provides insights on how to design antibodies with a desired functional effect, including those with neutralizing activity against bacterial and viral cell attachment proteins.


Asunto(s)
Adhesinas de Escherichia coli , Anticuerpos Neutralizantes , Proteínas Fimbrias , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/inmunología , Regulación Alostérica , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Proteínas Fimbrias/química , Proteínas Fimbrias/inmunología , Conformación Proteica
10.
Elife ; 112022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35881547

RESUMEN

A key attribute of persistent or recurring bacterial infections is the ability of the pathogen to evade the host's immune response. Many Enterobacteriaceae express type 1 pili, a pre-adapted virulence trait, to invade host epithelial cells and establish persistent infections. However, the molecular mechanisms and strategies by which bacteria actively circumvent the immune response of the host remain poorly understood. Here, we identified CD14, the major co-receptor for lipopolysaccharide detection, on mouse dendritic cells (DCs) as a binding partner of FimH, the protein located at the tip of the type 1 pilus of Escherichia coli. The FimH amino acids involved in CD14 binding are highly conserved across pathogenic and non-pathogenic strains. Binding of the pathogenic strain CFT073 to CD14 reduced DC migration by overactivation of integrins and blunted expression of co-stimulatory molecules by overactivating the NFAT (nuclear factor of activated T-cells) pathway, both rate-limiting factors of T cell activation. This response was binary at the single-cell level, but averaged in larger populations exposed to both piliated and non-piliated pathogens, presumably via the exchange of immunomodulatory cytokines. While defining an active molecular mechanism of immune evasion by pathogens, the interaction between FimH and CD14 represents a potential target to interfere with persistent and recurrent infections, such as urinary tract infections or Crohn's disease.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli Uropatógena , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/metabolismo , Animales , Infecciones por Escherichia coli/microbiología , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Inmunidad , Ratones , Escherichia coli Uropatógena/fisiología
11.
J Mol Biol ; 434(17): 167681, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35697293

RESUMEN

The FimH protein of Escherichia coli is a model two-domain adhesin that is able to mediate an allosteric catch bond mechanism of bacterial cell attachment, where the mannose-binding lectin domain switches from an 'inactive' conformation with fast binding to mannose to an 'active' conformation with slow detachment from mannose. Because mechanical tensile force favors separation of the domains and, thus, FimH activation, it has been thought that the catch bonds can only be manifested in a fluidic shear-dependent mode of adhesion. Here, we used recombinant FimH variants with a weakened inter-domain interaction and show that a fast and sustained allosteric activation of FimH can also occur under static, non-shear conditions. Moreover, it appears that lectin domain conformational activation happens intrinsically at a constant rate, independently from its ability to interact with the pilin domain or mannose. However, the latter two factors control the rate of FimH deactivation. Thus, the allosteric catch bond mechanism can be a much broader phenomenon involved in both fast and strong cell-pathogen attachments under a broad range of hydrodynamic conditions. This concept that allostery can enable more effective receptor-ligand interactions is fundamentally different from the conventional wisdom that allostery provides a mechanism to turn binding off under specific conditions.


Asunto(s)
Adhesinas de Escherichia coli , Adhesión Bacteriana , Escherichia coli , Proteínas Fimbrias , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/fisiología , Regulación Alostérica , Adhesión Bacteriana/fisiología , Escherichia coli/fisiología , Proteínas Fimbrias/química , Proteínas Fimbrias/genética , Manosa/metabolismo , Dominios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Resistencia al Corte
12.
NPJ Biofilms Microbiomes ; 8(1): 20, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35396507

RESUMEN

The formation of aggregates and biofilms enhances bacterial colonisation and infection progression by affording protection from antibiotics and host immune factors. Despite these advantages there is a trade-off, whereby bacterial dissemination is reduced. As such, biofilm development needs to be controlled to suit adaptation to different environments. Here we investigate members from one of largest groups of bacterial adhesins, the autotransporters, for their critical role in the assembly of bacterial aggregates and biofilms. We describe the structural and functional characterisation of autotransporter Ag43 variants from different Escherichia coli pathotypes. We show that specific interactions between amino acids on the contacting interfaces of adjacent Ag43 proteins drives a common mode of trans-association that leads to cell clumping. Furthermore, subtle variation of these interactions alters aggregation kinetics and the degree of compacting within cell clusters. Together, our structure-function investigation reveals an underlying molecular basis for variations in the density of bacterial communities.


Asunto(s)
Adhesinas de Escherichia coli , Proteínas de Escherichia coli , Adhesinas de Escherichia coli/química , Adhesión Bacteriana , Biopelículas , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo
13.
Biochem Biophys Res Commun ; 591: 37-43, 2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-34995984

RESUMEN

Antigen 43 is a surface-displayed autotransporter protein that mediates bacterial self-association and pathogenicity. The quality control factors that facilitate Ag43 crossing the periplasm and inserting into the outer membrane remain enigmatic, mostly because Ag43 is phase variable and associated with heterologous phenotypes, which obscures the mutational effects of potential quality control factors. Here, we describe a screening method that allowed us to isolate a subpopulation of Escherichia coli that consistently displays an Ag43-mediated autoaggregation phenotype. Based on this subpopulation, we analyzed how disruptions of known periplasmic chaperones affect Ag43 biogenesis. We found that only the disruption of surA reduced Ag43 levels and abolished the autoaggregation phenotype of cells, but surA disruption did not affect the phase-variable expression of agn43. Using purified proteins, we showed that SurA effectively protected the ß-barrel domain of Ag43 from aggregation. In contrast, the previously reported Ag43 biogenesis factor OsmY showed weak chaperoning effects on Ag43 only in the absence of SurA. Our results shed light on the roles of different periplasmic chaperones in Ag43 biogenesis and provide a methodology applicable to the study of other phase-variable proteins.


Asunto(s)
Adhesinas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Chaperonas Moleculares/metabolismo , Periplasma/metabolismo , Sistemas de Secreción Tipo V/metabolismo , Adhesinas de Escherichia coli/química , Proteínas Portadoras/metabolismo , Proteínas de Escherichia coli/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Fenotipo , Estructura Secundaria de Proteína
14.
J Comput Chem ; 42(21): 1526-1534, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-33982793

RESUMEN

MonteCarbo is an open-source software to construct simple 5-, 6-, and 7-membered ring multifunctionalized monosaccharides and nucleobases and dock them into the active site of carbohydrate-active enzymes. The core bash script executes simple orders to generate the Z-matrix of the neutral molecule of interest. After that, a Fortran90 code based on a pseudo-random number generator (Monte Carlo method) is executed to assign dihedral angles to the different rotamers present in the structure (ring and rotating functional groups). The program also has a generalized internal coordinates (GIC) implementation of the Cremer and Pople puckering coordinates ring. Once the structures are generated and optimized, a second code is ready to execute in serial the docking of multiple conformers in the active site of a wide family of enzymes.


Asunto(s)
Adhesinas de Escherichia coli/química , Carbohidratos/química , Proteínas Fimbrias/química , Simulación del Acoplamiento Molecular , Monosacáridos/química , Programas Informáticos , Adhesinas de Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Método de Montecarlo
15.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34011607

RESUMEN

Escherichia coli express adhesion pili that mediate attachment to host cell surfaces and are exposed to body fluids in the urinary and gastrointestinal tracts. Pilin subunits are organized into helical polymers, with a tip adhesin for specific host binding. Pili can elastically unwind when exposed to fluid flow forces, reducing the adhesin load, thereby facilitating sustained attachment. Here we investigate biophysical and structural differences of pili commonly expressed on bacteria that inhabit the urinary and intestinal tracts. Optical tweezers measurements reveal that class 1a pili of uropathogenic E. coli (UPEC), as well as class 1b of enterotoxigenic E. coli (ETEC), undergo an additional conformational change beyond pilus unwinding, providing significantly more elasticity to their structure than ETEC class 5 pili. Examining structural and steered molecular dynamics simulation data, we find that this difference in class 1 pili subunit behavior originates from an α-helical motif that can unfold when exposed to force. A disulfide bond cross-linking ß-strands in class 1 pili stabilizes subunits, allowing them to tolerate higher forces than class 5 pili that lack this covalent bond. We suggest that these extra contributions to pilus resiliency are relevant for the UPEC niche, since resident bacteria are exposed to stronger, more transient drag forces compared to those experienced by ETEC bacteria in the mucosa of the intestinal tract. Interestingly, class 1b ETEC pili include the same structural features seen in UPEC pili, while requiring lower unwinding forces that are more similar to those of class 5 ETEC pili.


Asunto(s)
Adhesinas de Escherichia coli/química , Escherichia coli Enterotoxigénica/ultraestructura , Proteínas Fimbrias/química , Fimbrias Bacterianas/ultraestructura , Escherichia coli Uropatógena/ultraestructura , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/metabolismo , Adhesión Bacteriana , Sitios de Unión , Fenómenos Biomecánicos , Cisteína/química , Cisteína/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Escherichia coli Enterotoxigénica/genética , Escherichia coli Enterotoxigénica/metabolismo , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Expresión Génica , Cinética , Simulación de Dinámica Molecular , Pinzas Ópticas , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Termodinámica , Escherichia coli Uropatógena/genética , Escherichia coli Uropatógena/metabolismo
16.
Molecules ; 26(3)2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33499241

RESUMEN

The Food and Drug Administration (FDA) approved a new class of anti-diabetic medication (a sodium-glucose co-transporter 2 (SGLT2) inhibitor) in 2013. However, SGLT2 inhibitor drugs are under evaluation due to their associative side effects, such as urinary tract and genital infection, urinary discomfort, diabetic ketosis, and kidney problems. Even clinicians have difficulty in recommending it to diabetic patients due to the increased probability of urinary tract infection. In our study, we selected natural SGLT2 inhibitors, namely acerogenin B, formononetin, (-)-kurarinone, (+)-pteryxin, and quinidine, to explore their potential against an emerging uropathogenic bacterial therapeutic target, i.e., FimH. FimH plays a critical role in the colonization of uropathogenic bacteria on the urinary tract surface. Thus, FimH antagonists show promising effects against uropathogenic bacterial strains via their targeting of FimH's adherence mechanism with less chance of resistance. The molecular docking results showed that, among natural SGLT2 inhibitors, formononetin, (+)-pteryxin, and quinidine have a strong interaction with FimH proteins, with binding energy (∆G) and inhibition constant (ki) values of -5.65 kcal/mol and 71.95 µM, -5.50 kcal/mol and 92.97 µM, and -5.70 kcal/mol and 66.40 µM, respectively. These interactions were better than those of the positive control heptyl α-d-mannopyranoside and far better than those of the SGLT2 inhibitor drug canagliflozin. Furthermore, a 50 ns molecular dynamics simulation was conducted to optimize the interaction, and the resulting complexes were found to be stable. Physicochemical property assessments predicted little toxicity and good drug-likeness properties for these three compounds. Therefore, formononetin, (+)-pteryxin, and quinidine can be proposed as promising SGLT2 inhibitors drugs, with add-on FimH inhibition potential that might reduce the probability of uropathogenic side effects.


Asunto(s)
Adhesinas de Escherichia coli/efectos de los fármacos , Infecciones por Escherichia coli/prevención & control , Proteínas Fimbrias/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/efectos adversos , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Infecciones Urinarias/prevención & control , Escherichia coli Uropatógena/efectos de los fármacos , Adhesinas de Escherichia coli/química , Biología Computacional , Simulación por Computador , Cumarinas/química , Cumarinas/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Infecciones por Escherichia coli/etiología , Proteínas Fimbrias/química , Humanos , Isoflavonas/química , Isoflavonas/farmacología , Simulación del Acoplamiento Molecular , Quinidina/química , Quinidina/farmacología , Transportador 2 de Sodio-Glucosa/química , Inhibidores del Cotransportador de Sodio-Glucosa 2/química , Infecciones Urinarias/etiología , Escherichia coli Uropatógena/patogenicidad
17.
Proteins ; 89(3): 276-288, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32989832

RESUMEN

Allostery governing two conformational states is one of the proposed mechanisms for catch-bond behavior in adhesive proteins. In FimH, a catch-bond protein expressed by pathogenic bacteria, separation of two domains disrupts inhibition by the pilin domain. Thus, tensile force can induce a conformational change in the lectin domain, from an inactive state to an active state with high affinity. To better understand allosteric inhibition in two-domain FimH (H2 inactive), we use molecular dynamics simulations to study the lectin domain alone, which has high affinity (HL active), and also the lectin domain stabilized in the low-affinity conformation by an Arg-60-Pro mutation (HL mutant). Because ligand-binding induces an allostery-like conformational change in HL mutant, this more experimentally tractable version has been proposed as a "minimal model" for FimH. We find that HL mutant has larger backbone fluctuations than both H2 inactive and HL active, at the binding pocket and allosteric interdomain region. We use an internal coordinate system of dihedral angles to identify protein regions with differences in backbone and side chain dynamics beyond the putative allosteric pathway sites. By characterizing HL mutant dynamics for the first time, we provide additional insight into the transmission of allosteric information across the lectin domain and build upon structural and thermodynamic data in the literature to further support the use of HL mutant as a "minimal model." Understanding how to alter protein dynamics to prevent the allosteric conformational change may guide drug development to prevent infection by blocking FimH adhesion.


Asunto(s)
Adhesinas de Escherichia coli , Proteínas Fimbrias , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/metabolismo , Sitio Alostérico , Proteínas Fimbrias/química , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Simulación de Dinámica Molecular , Mutación/genética , Conformación Proteica , Dominios Proteicos , Estabilidad Proteica , Termodinámica
18.
Biotechnol Bioeng ; 117(12): 3699-3711, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32827316

RESUMEN

We report a first of its kind functional cell surface display of nucleic acid polymerase and its directed evolution to efficiently incorporate 2'-O-methyl nucleotide triphosphates (2'-OMe-NTPs). In the development of polymerase cell surface display, two autotransporter proteins (Escherichia coli adhesin involved in diffuse adherence and Pseudomonas aeruginosa esterase A [EstA]) were employed to transport and anchor the 68-kDa Klenow fragment (KF) of E. coli DNA polymerase I on the surface of E. coli. The localization and function of the displayed KF were verified by analysis of cell outer membrane fractions, immunostaining, and fluorometric detection of synthesized DNA products. The EstA cell surface display system was applied to evolve KF for the incorporation of 2'-OMe-NTPs and a KF variant with a 50.7-fold increased ability to successively incorporate 2'-OMe-NTPs was discovered. Expanding the scope of cell-surface displayable proteins to the realm of polymerases provides a novel screening tool for tailoring polymerases to diverse application demands in a polymerase chain reaction and sequencing-based biotechnological and medical applications. Especially, cell surface display enables novel polymerase screening strategies in which the heat-lysis step is bypassed and thus allows the screening of mesophilic polymerases with broad application potentials ranging from diagnostics and DNA sequencing to replication of synthetic genetic polymers.


Asunto(s)
Adhesinas de Escherichia coli/química , Proteínas Bacterianas/química , Hidrolasas de Éster Carboxílico/química , ADN Polimerasa I/química , ADN Bacteriano/química , Evolución Molecular Dirigida , Escherichia coli/química , Pseudomonas aeruginosa/química
19.
Proteins ; 88(4): 593-603, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31622514

RESUMEN

FimH is a bacterial adhesin protein located at the tip of Escherichia coli fimbria that functions to adhere bacteria to host cells. Thus, FimH is a critical factor in bacterial infections such as urinary tract infections and is of interest in drug development. It is also involved in vaccine development and as a model for understanding shear-enhanced catch bond cell adhesion. To date, over 60 structures have been deposited in the Protein Data Bank showing interactions between FimH and mannose ligands, potential inhibitors, and other fimbrial proteins. In addition to providing insights about ligand recognition and fimbrial assembly, these structures provide insights into conformational changes in the two domains of FimH that are critical for its function. To gain further insights into these structural changes, we have superposed FimH's mannose binding lectin domain in all these structures and categorized the structures into five groups of lectin domain conformers using RMSD as a metric. Many structures also include the pilin domain, which anchors FimH to the fimbriae and regulates the conformation and function of the lectin domain. For these structures, we have also compared the relative orientations of the two domains. These structural analyses enhance our understanding of the conformational changes associated with FimH ligand binding and domain-domain interactions, including its catch bond behavior through allosteric action of force in bacterial adhesion.


Asunto(s)
Adhesinas de Escherichia coli/química , Escherichia coli/química , Proteínas Fimbrias/química , Fimbrias Bacterianas/química , Lectinas/química , Manosa/química , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/metabolismo , Regulación Alostérica , Adhesión Bacteriana , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Lectinas/genética , Lectinas/metabolismo , Ligandos , Manosa/genética , Manosa/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
20.
Soft Matter ; 16(4): 907-913, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31854427

RESUMEN

Biological systems have evolved sophisticated molecular assemblies capable of exquisite molecular recognition across length scales ranging from angstroms to microns. For instance, the self-organization of glycolipids and glycoproteins on cell membranes allows for molecular recognition of a diversity of ligands ranging from small molecules and proteins to viruses and whole cells. A distinguishing feature of these 2D surfaces is they achieve exceptional binding selectivity and avidity by exploiting multivalent binding interactions. Here we develop a 2D ligand display platform based on peptoid nanosheets that mimics the structure and function of the cell membrane. A variety of small-molecule lipid-conjugates were co-assembled with the peptoid chains to create a diversity of functionalized nanosheet bilayers with varying display densities. The functional heads of the lipids were shown to be surface-exposed, and the carbon tails immobilized into the hydrophobic interior. We demonstrate that saccharide-functionalized nanosheets (e.g., made from globotriaosylsphingosine or 1,2-dipalmitoyl-sn-glycero-3-phospho((ethyl-1',2',3'-triazole)triethyleneglycolmannose)) can have very diverse binding properties, exhibiting specific binding to multivalent proteins as well as to intact bacterial cells. Analysis of sugar display densities revealed that Shiga toxin 1 subunit B (a pentameric protein) and FimH-expressing Escherichia coli (E. coli) bind through the cooperative binding behavior of multiple carbohydrates. The ability to readily incorporate and display a wide variety of lipidated cargo on the surface of peptoid nanosheets makes this a convenient route to soluble, cell-surface mimetic materials. These materials hold great promise for drug screening, biosensing, bioremediation, and as a means to combat pathogens by direct physical binding through a well-defined, multivalent 2D material.


Asunto(s)
Carbohidratos/química , Lípidos/química , Nanoestructuras/química , Peptoides/química , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/genética , Biomimética , Escherichia coli/genética , Proteínas Fimbrias/química , Proteínas Fimbrias/genética , Interacciones Hidrofóbicas e Hidrofílicas , Toxina Shiga II/química , Toxina Shiga II/genética , Azúcares/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...