Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(37): e2208540119, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36070343

RESUMEN

Diversity Oriented Clicking (DOC) is a discovery method geared toward the rapid synthesis of functional libraries. It combines the best attributes of both classical and modern click chemistries. DOC strategies center upon the chemical diversification of core "SuFExable" hubs-exemplified by 2-Substituted-Alkynyl-1-Sulfonyl Fluorides (SASFs)-enabling the modular assembly of compounds through multiple reaction pathways. We report here a range of stereoselective Michael-type addition pathways from SASF hubs including reactions with secondary amines, carboxylates, 1H-1,2,3-triazole, and halides. These high yielding conjugate addition pathways deliver unprecedented ß-substituted alkenyl sulfonyl fluorides as single isomers with minimal purification, greatly enriching the repertoire of DOC and holding true to the fundamentals of modular click chemistry. Further, we demonstrate the potential for biological function - a key objective of click chemistry - of this family of SASF-derived molecules as covalent inhibitors of human neutrophil elastase.


Asunto(s)
Química Clic , Fluoruros , Elastasa de Leucocito , Proteínas Inhibidoras de Proteinasas Secretoras , Ácidos Sulfínicos , Química Clic/métodos , Fluoruros/síntesis química , Fluoruros/química , Fluoruros/farmacología , Humanos , Elastasa de Leucocito/antagonistas & inhibidores , Proteínas Inhibidoras de Proteinasas Secretoras/síntesis química , Proteínas Inhibidoras de Proteinasas Secretoras/química , Proteínas Inhibidoras de Proteinasas Secretoras/farmacología , Ácidos Sulfínicos/síntesis química , Ácidos Sulfínicos/química , Ácidos Sulfínicos/farmacología
2.
ACS Cent Sci ; 7(9): 1508-1515, 2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34584952

RESUMEN

Effector T cells comprise the cellular arm of the adaptive immune system and are essential for mounting immune responses against pathogens and cancer. To reach effector status, costimulation through CD28 is required. Here, we report that sialic acid-containing glycans on the surface of both T cells and APCs are alternative ligands of CD28 that compete with binding to its well-documented activatory ligand CD80 on the APC, resulting in attenuated costimulation. Removal of sialic acids enhances antigen-mediated activation of naïve T cells and also increases the revival of effector T cells made hypofunctional or exhausted via chronic viral infection. This occurs through a mechanism that is synergistic with antibody blockade of the inhibitory PD-1 axis. These results reveal a previously unrecognized role of sialic acid ligands in attenuation of CD28-mediated costimulation of T cells.

3.
Proc Natl Acad Sci U S A ; 117(31): 18431-18438, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32690700

RESUMEN

Influenza hemagglutinin (HA) glycoprotein is the primary surface antigen targeted by the host immune response and a focus for development of novel vaccines, broadly neutralizing antibodies (bnAbs), and therapeutics. HA enables viral entry into host cells via receptor binding and membrane fusion and is a validated target for drug discovery. However, to date, only a very few bona fide small molecules have been reported against the HA. To identity new antiviral lead candidates against the highly conserved fusion machinery in the HA stem, we synthesized a fluorescence-polarization probe based on a recently described neutralizing cyclic peptide P7 derived from the complementarity-determining region loops of human bnAbs FI6v3 and CR9114 against the HA stem. We then designed a robust binding assay compatible with high-throughput screening to identify molecules with low micromolar to nanomolar affinity to influenza A group 1 HAs. Our simple, low-cost, and efficient in vitro assay was used to screen H1/Puerto Rico/8/1934 (H1/PR8) HA trimer against ∼72,000 compounds. The crystal structure of H1/PR8 HA in complex with our best hit compound F0045(S) confirmed that it binds to pockets in the HA stem similar to bnAbs FI6v3 and CR9114, cyclic peptide P7, and small-molecule inhibitor JNJ4796. F0045 is enantioselective against a panel of group 1 HAs and F0045(S) exhibits in vitro neutralization activity against multiple H1N1 and H5N1 strains. Our assay, compound characterization, and small-molecule candidate should further stimulate the discovery and development of new compounds with unique chemical scaffolds and enhanced influenza antiviral capabilities.


Asunto(s)
Antivirales/farmacología , Evaluación Preclínica de Medicamentos/métodos , Polarización de Fluorescencia/métodos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/virología , Bibliotecas de Moléculas Pequeñas/farmacología , Antivirales/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Bibliotecas de Moléculas Pequeñas/química
4.
ACS Chem Biol ; 15(8): 2060-2069, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32662975

RESUMEN

Members of the CA class of cysteine proteases have multifaceted roles in physiology and virulence for many bacteria. Streptococcal pyrogenic exotoxin B (SpeB) is secreted by Streptococcus pyogenes and implicated in the pathogenesis of the bacterium through degradation of key human immune effector proteins. Here, we developed and characterized a clickable inhibitor, 2S-alkyne, based on X-ray crystallographic analysis and structure-activity relationships. Our SpeB probe showed irreversible enzyme inhibition in biochemical assays and labeled endogenous SpeB in cultured S. pyogenes supernatants. Importantly, application of 2S-alkyne decreased S. pyogenes survival in the presence of human neutrophils and supports the role of SpeB-mediated proteolysis as a mechanism to limit complement-mediated host defense. We posit that our SpeB inhibitor will be a useful chemical tool to regulate, label, and quantitate secreted cysteine proteases with SpeB-like activity in complex biological samples and a lead candidate for new therapeutics designed to sensitize S. pyogenes to host immune clearance.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Proteasas de Cisteína/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Streptococcus pyogenes/enzimología , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/química , Diseño de Fármacos , Conformación Proteica , Streptococcus pyogenes/patogenicidad , Relación Estructura-Actividad , Virulencia
5.
J Am Chem Soc ; 142(25): 10899-10904, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32479075

RESUMEN

Optimization of small-molecule probes or drugs is a synthetically lengthy, challenging, and resource-intensive process. Lack of automation and reliance on skilled medicinal chemists is cumbersome in both academic and industrial settings. Here, we demonstrate a high-throughput hit-to-lead process based on the biocompatible sulfur(VI) fluoride exchange (SuFEx) click chemistry. A high-throughput screening hit benzyl (cyanomethyl)carbamate (Ki = 8 µM) against a bacterial cysteine protease SpeB was modified with a SuFExable iminosulfur oxydifluoride [RN═S(O)F2] motif, rapidly diversified into 460 analogs in overnight reactions, and the products were directly screened to yield drug-like inhibitors with 480-fold higher potency (Ki = 18 nM). We showed that the improved molecule is active in a bacteria-host coculture. Since this SuFEx linkage reaction succeeds on picomole scale for direct screening, we anticipate our methodology can accelerate the development of robust biological probes and drug candidates.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Exotoxinas/antagonistas & inhibidores , Compuestos de Azufre/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Química Clic , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/toxicidad , Descubrimiento de Drogas , Exotoxinas/química , Exotoxinas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Jurkat , Microsomas Hepáticos/metabolismo , Prueba de Estudio Conceptual , Unión Proteica
6.
Proc Natl Acad Sci U S A ; 116(38): 18808-18814, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31484779

RESUMEN

Sulfur fluoride exchange (SuFEx) has emerged as the new generation of click chemistry. We report here a SuFEx-enabled, agnostic approach for the discovery and optimization of covalent inhibitors of human neutrophil elastase (hNE). Evaluation of our ever-growing collection of SuFExable compounds toward various biological assays unexpectedly revealed a selective and covalent hNE inhibitor: benzene-1,2-disulfonyl fluoride. Synthetic derivatization of the initial hit led to a more potent agent, 2-(fluorosulfonyl)phenyl fluorosulfate with IC50 0.24 µM and greater than 833-fold selectivity over the homologous neutrophil serine protease, cathepsin G. The optimized, yet simple benzenoid probe only modified active hNE and not its denatured form.


Asunto(s)
Fluoruros/química , Elastasa de Leucocito/antagonistas & inhibidores , Inhibidores de Serina Proteinasa/química , Compuestos de Azufre/química , Química Clic , Activación Enzimática/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Elastasa de Leucocito/química , Elastasa de Leucocito/metabolismo , Estructura Molecular , Unión Proteica , Pliegue de Proteína , Inhibidores de Serina Proteinasa/síntesis química , Inhibidores de Serina Proteinasa/farmacología , Ácidos Sulfínicos/síntesis química , Ácidos Sulfínicos/química , Ácidos Sulfínicos/farmacología
7.
Protein Sci ; 27(2): 509-522, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29114958

RESUMEN

Neutrophils contain high levels of chymotrypsin-like serine proteases (NSPs) within their azurophilic granules that have a multitude of functions within the immune system. In response, the pathogen Staphylococcus aureus has evolved three potent inhibitors (Eap, EapH1, and EapH2) that protect the bacterium as well as several of its secreted virulence factors from the degradative action of NSPs. We previously showed that these so-called EAP domain proteins represent a novel class of NSP inhibitors characterized by a non-covalent inhibitory mechanism and a distinct target specificity profile. Based upon high levels of structural homology amongst the EAP proteins and the NSPs, as well as supporting biochemical data, we predicted that the inhibited complex would be similar for all EAP/NSP pairs. However, we present here evidence that EapH1 and EapH2 bind the canonical NSP, Neutrophil Elastase (NE), in distinct orientations. We discovered that alteration of EapH1 residues at the EapH1/NE interface caused a dramatic loss of affinity and inhibition of NE, while mutation of equivalent positions in EapH2 had no effect on NE binding or inhibition. Surprisingly, mutation of residues in an altogether different region of EapH2 severely impacted both the NE binding and inhibitory properties of EapH2. Even though EapH1 and EapH2 bind and inhibit NE and a second NSP, Cathepsin G, equally well, neither of these proteins interacts with the structurally related, but non-proteolytic granule protein, azurocidin. These studies expand our understanding of EAP/NSP interactions and suggest that members of this immune evasion protein family are capable of diverse target recognition modes.


Asunto(s)
Elastasa de Leucocito/metabolismo , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/metabolismo , Staphylococcus aureus/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Línea Celular , Humanos , Evasión Inmune , Modelos Moleculares , Mutación , Unión Proteica , Dominios Proteicos , Inhibidores de Serina Proteinasa/genética , Staphylococcus aureus/enzimología
8.
Protein Sci ; 26(8): 1595-1608, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28512867

RESUMEN

The extracellular adherence protein (Eap) plays a crucial role in pathogenesis and survival of Staphylococcus aureus by inhibiting the classical and lectin pathways of complement. We have previously shown that Eap binds with nanomolar affinity to complement C4b and disrupts the initial interaction between C4b and C2, thereby inhibiting formation of the classical and lectin pathway C3 pro-convertase. Although an underlying mechanism has been identified, the structural basis for Eap binding to C4b is poorly understood. Here, we show that Eap domains 3 and 4 each contain a low-affinity, but saturable binding site for C4b. Taking advantage of the high lysine content of Eap, we used a zero-length crosslinking approach to map the Eap binding site to both the α'- and γ-chains of C4b. We also probed the C4b/Eap interface through a chemical footprinting approach involving lysine modification, proteolytic digestion, and mass spectrometry. This identified seven lysines in Eap that undergo changes in solvent exposure upon C4b binding. We found that simultaneous mutation of these lysines to either alanine or glutamate diminished C4b binding and complement inhibition by Eap. Together, our results provide insight into Eap recognition of C4b, and suggest that the repeating domains that comprise Eap are capable of multiple ligand-binding modes.


Asunto(s)
Proteínas Bacterianas/química , Complemento C4b/química , Lisina/química , Proteínas de Unión al ARN/química , Staphylococcus aureus/química , Alanina/química , Alanina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Carbodiimidas/química , Complemento C4b/genética , Complemento C4b/metabolismo , Vía Clásica del Complemento , Lectina de Unión a Manosa de la Vía del Complemento , Reactivos de Enlaces Cruzados/química , Cristalografía por Rayos X , Expresión Génica , Ácido Glutámico/química , Ácido Glutámico/metabolismo , Interacciones Huésped-Patógeno , Humanos , Lisina/metabolismo , Modelos Moleculares , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido
9.
Biomol NMR Assign ; 10(2): 301-5, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27372920

RESUMEN

The pathogenic bacterium Staphylococcus aureus has evolved to actively evade many aspects of the human innate immune system by expressing a series of secreted inhibitory proteins. Among these, the extracellular adherence protein (Eap) has been shown to inhibit the classical and lectin pathways of the complement system. By binding to complement component C4b, Eap is able to inhibit formation of the CP/LP C3 pro-convertase. Secreted full-length, mature Eap consists of four ~98 residue domains, all of which adopt a similar beta-grasp fold, and are connected through a short linker region. Through multiple biochemical approaches, it has been determined that the third and fourth domains of Eap are responsible for C4b binding. Here we report the backbone and side-chain resonance assignments of the 11.3 kDa fourth domain of Eap. The assignment data has been deposited in the BMRB database under the accession number 26726.


Asunto(s)
Proteínas Bacterianas/química , Resonancia Magnética Nuclear Biomolecular , Proteínas de Unión al ARN/química , Staphylococcus aureus , Dominios Proteicos
10.
Mol Immunol ; 67(2 Pt B): 193-205, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26052070

RESUMEN

The complement system plays a central role in a number of human inflammatory diseases, and there is a significant need for development of complement-directed therapies. The discovery of an arsenal of anti-complement proteins secreted by the pathogen Staphylococcus aureus brought with it the potential for harnessing the powerful inhibitory properties of these molecules. One such family of inhibitors, the SCINs, interact with a functional "hot-spot" on the surface of C3b. SCINs not only stabilize an inactive form of the alternative pathway (AP) C3 convertase (C3bBb), but also overlap the C3b binding site of complement factors B and H. Here we determined that a conserved Arg residue in SCINs is critical for function of full-length SCIN proteins. Despite this, we also found SCIN-specific differences in the contributions of other residues found at the C3b contact site, which suggested that a more diverse repertoire of residues might be able to recognize this region of C3b. To investigate this possibility, we conducted a phage display screen aimed at identifying SCIN-competitive 12-mer peptides. In total, seven unique sequences were identified and all exhibited direct C3b binding. A subset of these specifically inhibited the AP in assays of complement function. The mechanism of AP inhibition by these peptides was probed through surface plasmon resonance approaches, which revealed that six of the seven peptides disrupted C3bBb formation by interfering with factor B/C3b binding. To our knowledge this study has identified the first small molecules that retain inhibitory properties of larger staphylococcal immune evasion proteins.


Asunto(s)
Proteínas Bacterianas/metabolismo , Vía Alternativa del Complemento/efectos de los fármacos , Péptidos/farmacología , Staphylococcus aureus/metabolismo , Animales , Arginina/metabolismo , Unión Competitiva/efectos de los fármacos , C3 Convertasa de la Vía Alternativa del Complemento , Complemento C3b/metabolismo , Factor B del Complemento/metabolismo , Secuencia Conservada , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Biblioteca de Péptidos , Péptidos/química , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Conejos , Soluciones , Resonancia por Plasmón de Superficie
11.
J Immunol ; 193(12): 6161-6171, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25381436

RESUMEN

The pathogenic bacterium Staphylococcus aureus actively evades many aspects of human innate immunity by expressing a series of small inhibitory proteins. A number of these proteins inhibit the complement system, which labels bacteria for phagocytosis and generates inflammatory chemoattractants. Although the majority of staphylococcal complement inhibitors act on the alternative pathway to block the amplification loop, only a few proteins act on the initial recognition cascades that constitute the classical pathway (CP) and lectin pathway (LP). We screened a collection of recombinant, secreted staphylococcal proteins to determine whether S. aureus produces other molecules that inhibit the CP and/or LP. Using this approach, we identified the extracellular adherence protein (Eap) as a potent, specific inhibitor of both the CP and LP. We found that Eap blocked CP/LP-dependent activation of C3, but not C4, and that Eap likewise inhibited deposition of C3b on the surface of S. aureus cells. In turn, this significantly diminished the extent of S. aureus opsonophagocytosis and killing by neutrophils. This combination of functional properties suggested that Eap acts specifically at the level of the CP/LP C3 convertase (C4b2a). Indeed, we demonstrated a direct, nanomolar-affinity interaction of Eap with C4b. Eap binding to C4b inhibited binding of both full-length C2 and its C2b fragment, which indicated that Eap disrupts formation of the CP/LP C3 proconvertase (C4b2). As a whole, our results demonstrate that S. aureus inhibits two initiation routes of complement by expression of the Eap protein, and thereby define a novel mechanism of immune evasion.


Asunto(s)
Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , C3 Convertasa de la Vía Alternativa del Complemento/antagonistas & inhibidores , Vía Clásica del Complemento/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/metabolismo , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo , Proteínas Bacterianas/química , Sitios de Unión , Complemento C2/inmunología , Complemento C2/metabolismo , Complemento C3b/inmunología , Complemento C3b/metabolismo , Complemento C4b/inmunología , Complemento C4b/metabolismo , Citotoxicidad Inmunológica , Humanos , Modelos Inmunológicos , Neutrófilos/inmunología , Fagocitosis/inmunología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/química , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...