Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Front Immunol ; 14: 1109759, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37720229

RESUMEN

Introduction: Mucosal-associated invariant T (MAIT) cells are a population of innate-like T cells, which mediate host immunity to microbial infection by recognizing metabolite antigens derived from microbial riboflavin synthesis presented by the MHC-I-related protein 1 (MR1). Namely, the potent MAIT cell antigens, 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU) and 5-(2-oxoethylideneamino)-6-D-ribitylaminouracil (5-OE-RU), form via the condensation of the riboflavin precursor 5-amino-6-D-ribitylaminouracil (5-A-RU) with the reactive carbonyl species (RCS) methylglyoxal (MG) and glyoxal (G), respectively. Although MAIT cells are abundant in humans, they are rare in mice, and increasing their abundance using expansion protocols with antigen and adjuvant has been shown to facilitate their study in mouse models of infection and disease. Methods: Here, we outline three methods to increase the abundance of MAIT cells in C57BL/6 mice using a combination of inflammatory stimuli, 5-A-RU and MG. Results: Our data demonstrate that the administration of synthetic 5-A-RU in combination with one of three different inflammatory stimuli is sufficient to increase the frequency and absolute numbers of MAIT cells in C57BL/6 mice. The resultant boosted MAIT cells are functional and can provide protection against a lethal infection of Legionella longbeachae. Conclusion: These results provide alternative methods for expanding MAIT cells with high doses of commercially available 5-A-RU (± MG) in the presence of various danger signals.


Asunto(s)
Células T Invariantes Asociadas a Mucosa , Humanos , Animales , Ratones , Ratones Endogámicos C57BL , Adyuvantes Inmunológicos , Piruvaldehído , Riboflavina
2.
Front Immunol ; 12: 627173, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33777010

RESUMEN

Mucosal-associated invariant T (MAIT) cells are a population of innate-like T cells that utilize a semi-invariant T cell receptor (TCR) α chain and are restricted by the highly conserved antigen presenting molecule MR1. MR1 presents microbial riboflavin biosynthesis derived metabolites produced by bacteria and fungi. Consistent with their ability to sense ligands derived from bacterial sources, MAIT cells have been associated with the immune response to a variety of bacterial infections, such as Mycobacterium spp., Salmonella spp. and Escherichia coli. To date, MAIT cells have been studied in humans, non-human primates and mice. However, they have only been putatively identified in cattle by PCR based methods; no phenotypic or functional analyses have been performed. Here, we identified a MAIT cell population in cattle utilizing MR1 tetramers and high-throughput TCR sequencing. Phenotypic analysis of cattle MAIT cells revealed features highly analogous to those of MAIT cells in humans and mice, including expression of an orthologous TRAV1-TRAJ33 TCR α chain, an effector memory phenotype irrespective of tissue localization, and expression of the transcription factors PLZF and EOMES. We determined the frequency of MAIT cells in peripheral blood and multiple tissues, finding that cattle MAIT cells are enriched in mucosal tissues as well as in the mesenteric lymph node. Cattle MAIT cells were responsive to stimulation by 5-OP-RU and riboflavin biosynthesis competent bacteria in vitro. Furthermore, MAIT cells in milk increased in frequency in cows with mastitis. Following challenge with virulent Mycobacterium bovis, a causative agent of bovine tuberculosis and a zoonosis, peripheral blood MAIT cells expressed higher levels of perforin. Thus, MAIT cells are implicated in the immune response to two major bacterial infections in cattle. These data suggest that MAIT cells are functionally highly conserved and that cattle are an excellent large animal model to study the role of MAIT cells in important zoonotic infections.


Asunto(s)
Infecciones Bacterianas/inmunología , Bovinos/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Animales , Citocinas/farmacología , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Masculino , Ratones , Antígenos de Histocompatibilidad Menor/inmunología , Fenotipo , Ribitol/análogos & derivados , Ribitol/farmacología , Uracilo/análogos & derivados , Uracilo/farmacología
3.
J Immunol ; 206(7): 1425-1435, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33597151

RESUMEN

Mucosal-associated invariant T (MAIT) cells are an innate-like population of unconventional T cells that respond rapidly to microbial metabolite Ags or cytokine stimulation. Because of this reactivity and surface expression of CD45RO+, CD45RA-, and CD127+, they are described as effector memory cells. Yet, there is heterogeneity in MAIT cell effector response. It is unclear what factors control MAIT cell effector capacity, whether it is fixed or can be modified and if this differs based on whether activation is TCR dependent or independent. To address this, we have taken a systematic approach to examine human MAIT cell effector capacity across healthy individuals in response to ligand and cytokine stimulation. We demonstrate the heterogenous nature of MAIT cell effector capacity and that the ability to produce an effector response is not directly attributable to TCR clonotype or coreceptor expression. Global gene transcription analysis revealed that the MAIT cell effector capacity produced in response to TCR stimulation is associated with increased expression of the epigenetic regulator lysine demethylase 6B (KDM6B). Addition of a KDM6B inhibitor did not alter MAIT cell effector function to Ag or cytokine stimulation. However, addition of the KDM6B cofactor α-ketoglutarate greatly enhanced MAIT cell effector capacity to TCR-dependent stimulation in a partially KDM6B-dependent manner. These results demonstrate that the TCR-dependent effector response of MAIT cells is epigenetically regulated and dependent on the availability of metabolic cofactors.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/metabolismo , Ácidos Cetoglutáricos/metabolismo , Células T Invariantes Asociadas a Mucosa/inmunología , Células Cultivadas , Citocinas/metabolismo , Epigénesis Genética , Humanos , Inmunidad Innata , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/metabolismo
4.
Curr Protoc Immunol ; 127(1): e90, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31763790

RESUMEN

Mucosal-associated invariant T (MAIT) cells are a subset of unconventional T cells restricted by the major histocompatibility complex (MHC) class I-like molecule MHC-related protein 1 (MR1). MAIT cells are found throughout the body, especially in human blood and liver. Unlike conventional T cells, which are stimulated by peptide antigens presented by MHC molecules, MAIT cells recognize metabolite antigens derived from an intermediate in the microbial biosynthesis of riboflavin. MAIT cells mediate protective immunity to infections by riboflavin-producing microbes via the production of cytokines and cytotoxicity. The discovery of stimulating MAIT cell antigens allowed for the development of an analytical tool, the MR1 tetramer, that binds specifically to the MAIT T cell receptor (TCR) and is becoming the gold standard for identification of MAIT cells by flow cytometry. This article describes protocols to characterize the phenotype of human MAIT cells in blood and tissues by flow cytometry using fluorescently labeled human MR1 tetramers alongside antibodies specific for MAIT cell markers. © 2019 by John Wiley & Sons, Inc. The main protocols include: Basic Protocol 1: Determining the frequency and steady-state surface phenotype of human MAIT cells Basic Protocol 2: Determining the activation phenotype of human MAIT cells in blood Basic Protocol 3: Characterizing MAIT cell TCRs using TCR-positive reporter cell lines Alternate protocols are provided for determining the absolute number, transcription factor phenotype, and TCR usage of human MAIT cells; and determining activation phenotype by staining for intracellular markers, measuring secreted cytokines, and measuring fluorescent dye dilution due to proliferation. Additional methods are provided for determining the capacity of MAIT cells to produce cytokine independently of antigen using plate-bound or bead-immobilized CD3/CD28 stimulation; and determining the MR1-Ag dependence of MAIT cell activation using MR1-blocking antibody or competitive inhibition. For TCR-positive reporter cell lines, methods are also provided for evaluating the MAIT TCR-mediated MR1-Ag response, determining the capacity of the reporter lines to produce cytokine independently of antigen, determining the MR1-Ag dependence of the reporter lines, and evaluating the MR1-Ag response of the reporter lines using IL-2 secretion. Support Protocols describe the preparation of PBMCs from human blood, the preparation of single-cell suspensions from tissue, the isolation of MAIT cells by FACS and MACS, cloning MAIT TCRα and ß chain genes and MR1 genes for transduction, generating stably and transiently transfected cells lines, generating a stable MR1 knockout antigen-presenting cell line, and generating monocyte-derived dendritic cells.


Asunto(s)
Células T Invariantes Asociadas a Mucosa/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Citometría de Flujo , Humanos , Células T Invariantes Asociadas a Mucosa/citología , Fenotipo
5.
J Chem Inf Model ; 55(6): 1181-91, 2015 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-26000704

RESUMEN

Protease activated receptor 2 (PAR2) is an unusual G-protein coupled receptor (GPCR) involved in inflammation and metabolism. It is activated through cleavage of its N-terminus by proteases. The new N-terminus functions as a tethered ligand that folds back and intramolecularly activates PAR2, initiating multiple downstream signaling pathways. The only compounds reported to date to inhibit PAR2 activation are of moderate potency. Three structural models for PAR2 have been constructed based on sequence homology with known crystal structures for bovine rhodopsin, human ORL-1 (also called nociceptin/orphanin FQ receptor), and human PAR1. The three PAR2 model structures were compared and used to predict potential interactions with ligands. Virtual screening for ligands using the Chembridge database, and either ORL-1 or PAR1 derived PAR2 models led to identification of eight new small molecule PAR2 antagonists (IC50 10-100 µM). Notably, the most potent compound 1 (IC50 11 µM) was derived from the less homologous template protein, human ORL-1. The results suggest that virtual screening against multiple homology models of the same GPCR can produce structurally diverse antagonists and that this may be desirable even when some models have less sequence homology with the target protein.


Asunto(s)
Descubrimiento de Drogas/métodos , Simulación del Acoplamiento Molecular , Receptor PAR-2/antagonistas & inhibidores , Receptor PAR-2/química , Homología de Secuencia de Aminoácido , Animales , Sitios de Unión , Bovinos , Membrana Celular/metabolismo , Bases de Datos de Proteínas , Evaluación Preclínica de Medicamentos , Células HT29 , Humanos , Ligandos , Estructura Terciaria de Proteína , Receptor PAR-2/metabolismo
6.
ACS Chem Biol ; 10(4): 957-64, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25603425

RESUMEN

The DsbA:DsbB redox machinery catalyzes disulfide bond formation in secreted proteins and is required for bacterial virulence factor assembly. Both enzymes have been identified as targets for antivirulence drugs. Here, we report synthetic analogues of ubiquinone (dimedone derivatives) that inhibit disulfide bond formation (IC50∼1 µM) catalyzed by E. coli DsbA:DsbB. The mechanism involves covalent modification of a single free cysteine leaving other cysteines unmodified. A vinylogous anhydride in each inhibitor is cleaved by the thiol, which becomes covalently modified to a thioester by a propionyl substituent. Cysteines and lysines on DsbA and DsbB and a nonredox enzyme were modified in a manner that implies some specificity. Moreover, human thioredoxin was not inhibited under the same conditions that inhibited EcDsbA. This proof of concept work uses small molecules that target specific cysteines to validate the DsbA and DsbB dual enzyme system as a viable and potentially druggable antivirulence target.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/química , Disulfuros/química , Proteínas de Escherichia coli/química , Proteínas de la Membrana/química , Proteína Disulfuro Isomerasas/química , Antibacterianos/síntesis química , Proteínas Bacterianas/metabolismo , Cisteína/química , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Proteínas de Escherichia coli/metabolismo , Humanos , Concentración 50 Inhibidora , Lisina/química , Proteínas de la Membrana/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Tiorredoxinas/antagonistas & inhibidores , Ubiquinona/análogos & derivados
7.
J Med Chem ; 57(20): 8459-70, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25259874

RESUMEN

The G-protein coupled receptor (C3aR) for human inflammatory protein complement C3a is an important component of immune, inflammatory, and metabolic diseases. A flexible compound (N2-[(2,2-diphenylethoxy)acetyl]-l-arginine, 4), known as a weak C3aR antagonist (IC50 µM), was transformed here into potent agonists (EC50 nM) of human macrophages (Ca(2+) release in HMDM) by incorporating aromatic heterocycles. Antagonists were also identified. A linear correlation between binding affinity for C3aR and calculated hydrogen-bond interaction energy of the heteroatom indicated that its hydrogen-bonding capacity influenced ligand affinity and function mediated by C3aR. Hydrogen-bond accepting heterocycles (e.g., imidazole) conferred the highest affinity and agonist potency (e.g., 21, EC50 24 nM, Ca(2+), HMDM) with comparable efficacy and immunostimulatory activity as that of C3a in activating human macrophages (Ca(2+), IL1ß, TNFα, CCL3). These potent and selective modulators of C3aR, inactivated by a C3aR antagonist, are stable C3a surrogates for interrogating roles for C3aR in physiology and disease.


Asunto(s)
Compuestos Heterocíclicos/química , Receptores de Complemento/agonistas , Receptores de Complemento/metabolismo , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Arginina/análogos & derivados , Arginina/química , Arginina/farmacología , Compuestos de Bencidrilo/química , Compuestos de Bencidrilo/farmacología , Calcio/metabolismo , Células Cultivadas , Técnicas de Química Sintética , Regulación de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Humanos , Enlace de Hidrógeno , Ligandos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Receptores de Complemento/antagonistas & inhibidores , Relación Estructura-Actividad
8.
Antiviral Res ; 84(3): 234-41, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19781577

RESUMEN

The flaviviruses comprise a large group of related viruses, many of which pose a significant global human health threat, most notably the dengue viruses (DENV), West Nile virus (WNV) and yellow fever virus (YFV). Flaviviruses enter host cells via fusion of the viral and cellular membranes, a process mediated by the major viral envelope protein E as it undergoes a low pH induced conformational change in the endosomal compartment of the host cell. This essential entry stage in the flavivirus life cycle provides an attractive target for the development of antiviral agents. We performed an in silico docking screen of the Maybridge chemical database within a previously described ligand binding pocket in the dengue E protein structure that is thought to play a key role in the conformational transitions that lead to membrane fusion. The biological activity of selected compounds identified from this screen revealed low micromolar antiviral potency against dengue virus for two of the compounds. Our results also provide the first evidence that compounds selected to bind to this ligand binding site on the flavivirus E protein abrogate fusion activity. Interestingly, one of these compounds also has antiviral activity against both WNV (kunjin strain) and YFV.


Asunto(s)
Antivirales/farmacología , Evaluación Preclínica de Medicamentos , Infecciones por Flavivirus/virología , Flavivirus/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Animales , Sitios de Unión , Chlorocebus aethiops , Virus del Dengue/química , Virus del Dengue/efectos de los fármacos , Virus del Dengue/fisiología , Flavivirus/química , Flavivirus/fisiología , Infecciones por Flavivirus/tratamiento farmacológico , Humanos , Unión Proteica , Células Vero , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos
9.
Chembiochem ; 4(2-3): 181-5, 2003 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-12616631

RESUMEN

Few reported inhibitors of secretory phospholipase A(2) enzymes truly inhibit the IIa human isoform (hnpsPLA(2)-IIa) noncovalently at submicromolar concentrations. Herein, the simple chiral precursor D-tyrosine was derivatised to give a series of potent new inhibitors of hnpsPLA(2)-IIa. A 2.2-A crystal structure shows an inhibitor bound in the active site of the enzyme, chelated to a Ca(2+) ion through carboxylate and amide oxygen atoms, H-bonded through an amide NH group to His48, with multiple hydrophobic contacts and a T-shaped aromatic-group-His6 interaction. Antiinflammatory activity is also demonstrated for two compounds administered orally to rats.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Inflamación/tratamiento farmacológico , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A/farmacología , Tirosina/síntesis química , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/prevención & control , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/química , Humanos , Inflamación/inducido químicamente , Inflamación/prevención & control , Fosfolipasas A2 , Ratas , Ratas Wistar , Relación Estructura-Actividad , Tirosina/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA