Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Biol Chem ; 298(2): 101463, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34864058

RESUMEN

Interleukin (IL)-22 is a cytokine that plays a critical role in intestinal epithelial homeostasis. Its downstream functions are mediated through interaction with the heterodimeric IL-22 receptor and subsequent activation of signal transducer and activator of transcription 3 (STAT3). IL-22 signaling can induce transcription of genes necessary for intestinal epithelial cell proliferation, tissue regeneration, tight junction fortification, and antimicrobial production. Recent studies have also implicated IL-22 signaling in the regulation of intestinal epithelial fucosylation in mice. However, whether IL-22 regulates intestinal fucosylation in human intestinal epithelial cells and the molecular mechanisms that govern this process are unknown. Here, in experiments performed in human cell lines and human-derived enteroids, we show that IL-22 signaling regulates expression of the B3GNT7 transcript, which encodes a ß1-3-N-acetylglucosaminyltransferase that can participate in the synthesis of poly-N-acetyllactosamine (polyLacNAc) chains. Additionally, we find that IL-22 signaling regulates levels of the α1-3-fucosylated Lewis X (Lex) blood group antigen, and that this glycan epitope is primarily displayed on O-glycosylated intestinal epithelial glycoproteins. Moreover, we show that increased expression of B3GNT7 alone is sufficient to promote increased display of Lex-decorated carbohydrate glycan structures primarily on O-glycosylated intestinal epithelial glycoproteins. Together, these data identify B3GNT7 as an intermediary in IL-22-dependent induction of fucosylation of glycoproteins and uncover a novel role for B3GNT7 in intestinal glycosylation.


Asunto(s)
Células Epiteliales , Glicoproteínas , Interleucinas , Mucosa Intestinal , N-Acetilglucosaminiltransferasas , Células Epiteliales/metabolismo , Glicoproteínas/metabolismo , Glicosilación , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , N-Acetilglucosaminiltransferasas/biosíntesis , N-Acetilglucosaminiltransferasas/metabolismo , Polisacáridos/metabolismo , Interleucina-22
2.
Nature ; 505(7481): 103-7, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24256734

RESUMEN

Human body-surface epithelia coexist in close association with complex bacterial communities and are protected by a variety of antibacterial proteins. C-type lectins of the RegIII family are bactericidal proteins that limit direct contact between bacteria and the intestinal epithelium and thus promote tolerance to the intestinal microbiota. RegIII lectins recognize their bacterial targets by binding peptidoglycan carbohydrate, but the mechanism by which they kill bacteria is unknown. Here we elucidate the mechanistic basis for RegIII bactericidal activity. We show that human RegIIIα (also known as HIP/PAP) binds membrane phospholipids and kills bacteria by forming a hexameric membrane-permeabilizing oligomeric pore. We derive a three-dimensional model of the RegIIIα pore by docking the RegIIIα crystal structure into a cryo-electron microscopic map of the pore complex, and show that the model accords with experimentally determined properties of the pore. Lipopolysaccharide inhibits RegIIIα pore-forming activity, explaining why RegIIIα is bactericidal for Gram-positive but not Gram-negative bacteria. Our findings identify C-type lectins as mediators of membrane attack in the mucosal immune system, and provide detailed insight into an antibacterial mechanism that promotes mutualism with the resident microbiota.


Asunto(s)
Antibacterianos/metabolismo , Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Intestinos/química , Lectinas Tipo C/metabolismo , Porinas/metabolismo , Antibacterianos/química , Antibacterianos/inmunología , Antibacterianos/farmacología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/química , Biomarcadores de Tumor/inmunología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Microscopía por Crioelectrón , Cristalografía por Rayos X , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/inmunología , Bacterias Gramnegativas/metabolismo , Humanos , Inmunidad Mucosa/efectos de los fármacos , Inmunidad Mucosa/inmunología , Intestinos/inmunología , Intestinos/microbiología , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/química , Lectinas Tipo C/inmunología , Lipopolisacáridos/farmacología , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/inmunología , Listeria monocytogenes/metabolismo , Viabilidad Microbiana/efectos de los fármacos , Modelos Moleculares , Proteínas Asociadas a Pancreatitis , Peptidoglicano/metabolismo , Fosfolípidos/metabolismo , Porinas/antagonistas & inhibidores , Porinas/química , Simbiosis
3.
Proc Natl Acad Sci U S A ; 114(42): 11027-11033, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-28973871

RESUMEN

The mammalian intestine is colonized by trillions of bacteria that perform essential metabolic functions for their hosts. The mutualistic nature of this relationship depends on maintaining spatial segregation between these bacteria and the intestinal epithelial surface. This segregation is achieved in part by the presence of a dense mucus layer at the epithelial surface and by the production of antimicrobial proteins that are secreted by epithelial cells into the mucus layer. Here, we show that resistin-like molecule ß (RELMß) is a bactericidal protein that limits contact between Gram-negative bacteria and the colonic epithelial surface. Mouse and human RELMß selectively killed Gram-negative bacteria by forming size-selective pores that permeabilized bacterial membranes. In mice lacking RELMß, Proteobacteria were present in the inner mucus layer and invaded mucosal tissues. Another RELM family member, human resistin, was also bactericidal, suggesting that bactericidal activity is a conserved function of the RELM family. Our findings thus identify the RELM family as a unique family of bactericidal proteins and show that RELMß promotes host-bacterial mutualism by regulating the spatial segregation between the microbiota and the intestinal epithelium.


Asunto(s)
Microbioma Gastrointestinal , Bacterias Gramnegativas , Hormonas Ectópicas/fisiología , Mucosa Intestinal/microbiología , Animales , Humanos , Inmunidad Innata , Péptidos y Proteínas de Señalización Intercelular , Mucosa Intestinal/inmunología , Metabolismo de los Lípidos , Ratones , Resistina/fisiología , Simbiosis
4.
Elife ; 122023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37159507

RESUMEN

Peristaltic movement of the intestine propels food down the length of the gastrointestinal tract to promote nutrient absorption. Interactions between intestinal macrophages and the enteric nervous system regulate gastrointestinal motility, yet we have an incomplete understanding of the molecular mediators of this crosstalk. Here, we identify complement component 1q (C1q) as a macrophage product that regulates gut motility. Macrophages were the predominant source of C1q in the mouse intestine and most extraintestinal tissues. Although C1q mediates the complement-mediated killing of bacteria in the bloodstream, we found that C1q was not essential for the immune defense of the intestine. Instead, C1q-expressing macrophages were located in the intestinal submucosal and myenteric plexuses where they were closely associated with enteric neurons and expressed surface markers characteristic of nerve-adjacent macrophages in other tissues. Mice with a macrophage-specific deletion of C1qa showed changes in enteric neuronal gene expression, increased neurogenic activity of peristalsis, and accelerated intestinal transit. Our findings identify C1q as a key regulator of gastrointestinal motility and provide enhanced insight into the crosstalk between macrophages and the enteric nervous system.


Asunto(s)
Complemento C1q , Sistema Nervioso Entérico , Ratones , Animales , Complemento C1q/metabolismo , Motilidad Gastrointestinal/fisiología , Macrófagos/metabolismo , Tracto Gastrointestinal
5.
Science ; 374(6568): eabe6723, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34735226

RESUMEN

A diverse group of antimicrobial proteins (AMPs) helps protect the mammalian intestine from varied microbial challenges. We show that small proline-rich protein 2A (SPRR2A) is an intestinal antibacterial protein that is phylogenetically unrelated to previously discovered mammalian AMPs. In this study, SPRR2A was expressed in Paneth cells and goblet cells and selectively killed Gram-positive bacteria by disrupting their membranes. SPRR2A shaped intestinal microbiota composition, restricted bacterial association with the intestinal surface, and protected against Listeria monocytogenes infection. SPRR2A differed from other intestinal AMPs in that it was induced by type 2 cytokines produced during helminth infection. Moreover, SPRR2A protected against helminth-induced bacterial invasion of intestinal tissue. Thus, SPRR2A is a distinctive AMP triggered by type 2 immunity that protects the intestinal barrier during helminth infection.


Asunto(s)
Proteínas Ricas en Prolina del Estrato Córneo/metabolismo , Microbioma Gastrointestinal , Bacterias Grampositivas/fisiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Nematospiroides dubius , Infecciones por Strongylida/inmunología , Animales , Carga Bacteriana , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Proteínas Ricas en Prolina del Estrato Córneo/genética , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Células Caliciformes/metabolismo , Humanos , Inmunidad Innata , Mucosa Intestinal/microbiología , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Ratones , Viabilidad Microbiana , Células de Paneth/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/microbiología
6.
Cell Host Microbe ; 25(6): 777-788.e8, 2019 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-31101494

RESUMEN

Vitamin A deficiency increases susceptibility to skin infection. However, the mechanisms by which vitamin A regulates skin immunity remain unclear. Here, we show that resistin-like molecule α (RELMα), a small secreted cysteine-rich protein, is expressed by epidermal keratinocytes and sebocytes and serves as an antimicrobial protein that is required for vitamin-A-dependent resistance to skin infection. RELMα was induced by microbiota colonization of the murine skin, was bactericidal in vitro, and was protected against bacterial infection of the skin in vivo. RELMα expression required dietary vitamin A and was induced by the therapeutic vitamin A analog isotretinoin, which protected against skin infection in a RELMα-dependent manner. The RELM family member Resistin was expressed in human skin, was induced by vitamin A analogs, and killed skin bacteria, indicating a conserved function for RELM proteins in skin innate immunity. Our findings provide insight into how vitamin A promotes resistance to skin infection.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Factores Inmunológicos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Enfermedades Cutáneas Bacterianas/prevención & control , Piel/inmunología , Vitamina A/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Ratones , Resistina/metabolismo , Enfermedades Cutáneas Bacterianas/inmunología , Activación Transcripcional/efectos de los fármacos
8.
Cell Host Microbe ; 18(4): 392-4, 2015 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-26468742

RESUMEN

Humans are home to complex communities of microbes that profoundly influence host physiology. Two recent papers, including Earle et al. (2015) in this issue of Cell Host & Microbe, contribute new experimental tools for visualizing and tracking bacteria within a host, promising to shed light on how microbes interact to influence host health.


Asunto(s)
Técnicas Bacteriológicas/métodos , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Microscopía Fluorescente/métodos , Imagen Óptica/métodos , Animales
9.
Mol Biosyst ; 7(7): 2114-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21597618

RESUMEN

Herein we describe the orientation of GST-tagged lectins on NHS-activated slides via a one-step deposition of the protein and a glutathione (GSH) scaffold. This technology overcomes the need for a premade GSH-surface to orient GST-tagged proteins, enabling us to rapidly expand the analytical capacity of lectin microarrays through addition of oriented lectins, while maintaining lectin diversity.


Asunto(s)
Glutatión Transferasa/metabolismo , Glicómica/métodos , Lectinas/metabolismo , Análisis por Matrices de Proteínas/métodos , Proteínas Recombinantes de Fusión/metabolismo , Animales , Bovinos , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Albúmina Sérica Bovina/metabolismo , Propiedades de Superficie
10.
Methods Mol Biol ; 723: 67-77, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21370060

RESUMEN

The cell surface is covered with a myriad of carbohydrates that form a complex matrix of oligosaccharides. Carbohydrate recognition plays critical roles in pathogenesis, trafficking, and differentiation. Lectin microarray technology presents a novel platform for the high-throughput analysis of these structurally diverse biopolymers. One drawback of this technology has been limitations imposed by the commercially available plant lectins used in the array. Not only are a majority of these plant-derived proteins glycosylated, which can complicate glycomic analysis, but they also differ in activity and availability. Our lab has recently introduced recombinant lectins to enhance the stability and scope of our lectin panel. Herein, we provide a detailed procedure for the expression of bacterially-derived lectins and their application to a recombinant lectin microarray.


Asunto(s)
Glicómica/métodos , Lectinas/metabolismo , Análisis por Matrices de Proteínas/métodos , Proteínas Recombinantes/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Línea Celular Tumoral , ADN Recombinante/genética , Ensayo de Inmunoadsorción Enzimática , Lectinas/genética , Lectinas/aislamiento & purificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA