Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sci Signal ; 17(827): eade3643, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38470955

RESUMEN

Activation of the endoplasmic reticulum (ER)-resident adaptor protein STING, a component of a cytosolic DNA-sensing pathway, induces the transcription of genes encoding type I interferons (IFNs) and other proinflammatory factors. Because STING is activated at the Golgi apparatus, control of the localization and activation of STING is important in stimulating antiviral and antitumor immune responses. Through a genome-wide CRISPR interference screen, we found that STING activation required the Golgi-resident protein ACBD3, which promotes the generation of phosphatidylinositol 4-phosphate (PI4P) at the trans-Golgi network, as well as other PI4P-associated proteins. Appropriate localization and activation of STING at the Golgi apparatus required ACBD3 and the PI4P-generating kinase PI4KB. In contrast, STING activation was enhanced when the lipid-shuttling protein OSBP, which removes PI4P from the Golgi apparatus, was inhibited by the US Food and Drug Administration-approved antifungal itraconazole. The increase in the abundance of STING-activating phospholipids at the trans-Golgi network resulted in the increased production of IFN-ß and other cytokines in THP-1 cells. Furthermore, a mutant STING that could not bind to PI4P failed to traffic from the ER to the Golgi apparatus in response to a STING agonist, whereas forced relocalization of STING to PI4P-enriched areas elicited STING activation in the absence of stimulation with a STING agonist. Thus, PI4P is critical for STING activation, and manipulating PI4P abundance may therapeutically modulate STING-dependent immune responses.


Asunto(s)
Aparato de Golgi , Fosfolípidos , Fosfolípidos/metabolismo , Aparato de Golgi/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
2.
J Cell Sci ; 137(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38345099

RESUMEN

Glycosylated mucin proteins contribute to the essential barrier function of the intestinal epithelium. The transmembrane mucin MUC13 is an abundant intestinal glycoprotein with important functions for mucosal maintenance that are not yet completely understood. We demonstrate that in human intestinal epithelial monolayers, MUC13 localized to both the apical surface and the tight junction (TJ) region on the lateral membrane. MUC13 deletion resulted in increased transepithelial resistance (TEER) and reduced translocation of small solutes. TEER buildup in ΔMUC13 cells could be prevented by addition of MLCK, ROCK or protein kinase C (PKC) inhibitors. The levels of TJ proteins including claudins and occludin were highly increased in membrane fractions of MUC13 knockout cells. Removal of the MUC13 cytoplasmic tail (CT) also altered TJ composition but did not affect TEER. The increased buildup of TJ complexes in ΔMUC13 and MUC13-ΔCT cells was dependent on PKC. The responsible PKC member might be PKCδ (or PRKCD) based on elevated protein levels in the absence of full-length MUC13. Our results demonstrate for the first time that a mucin protein can negatively regulate TJ function and stimulate intestinal barrier permeability.


Asunto(s)
Proteína Quinasa C , Proteínas de Uniones Estrechas , Humanos , Proteínas de Uniones Estrechas/metabolismo , Proteína Quinasa C/metabolismo , Intestinos , Mucosa Intestinal/metabolismo , Uniones Estrechas/metabolismo , Ocludina , Mucinas/metabolismo , Células Epiteliales/metabolismo
3.
Viruses ; 13(4)2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807137

RESUMEN

The trimeric hemagglutinin-esterase fusion protein (HEF) of influenza D virus (IDV) binds 9-O-acetylated sialic acid receptors, which are expressed in various host species. While cattle are the main reservoir for IDV, the viral genome has also been detected in domestic pigs. In addition, antibodies against IDV have been detected in other farm animals such as sheep, goats, and horses, and even in farmers working with IDV positive animals. Viruses belonging to various IDV clades circulate, but little is known about their differences in host and tissue tropism. Here we used recombinantly produced HEF proteins (HEF S57A) from the major clades D/Oklahoma (D/OK) and D/Oklahoma/660 (D/660) to study their host and tissue tropism and receptor interactions. To this end, we developed tissue microarrays (TMA) composed of respiratory tissues from various farm animals including cattle, domestic pigs, sheep, goats, and horses. Protein histochemical staining of farm animal respiratory tissue-microarrays with HEF proteins showed that cattle have receptors present over the entire respiratory tract while receptors are only present in the nasal and pharyngeal epithelium of pigs, sheep, goats, and horses. No differences in tropism for tissues and animals were observed between clades, while hemagglutination assays showed that D/OK has a 2-fold higher binding affinity than D/660 for receptors on red blood cells. The removal of O-acetylation from receptors via saponification treatment confirmed that receptor-binding of both clades was dependent on O-acetylated sialic acids.


Asunto(s)
Hemaglutininas Virales/metabolismo , Sistema Respiratorio/virología , Thogotovirus/fisiología , Análisis de Matrices Tisulares , Proteínas Virales de Fusión/metabolismo , Tropismo Viral , Acoplamiento Viral , Animales , Animales Domésticos/virología , Bovinos , Cabras , Hemaglutininas Virales/genética , Caballos , Interacciones Microbiota-Huesped , Proteínas Recombinantes/metabolismo , Ovinos , Ácidos Siálicos/metabolismo , Porcinos , Thogotovirus/química , Thogotovirus/genética , Proteínas Virales de Fusión/genética
4.
mBio ; 12(2)2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824202

RESUMEN

At the intestinal host-microbe interface, the transmembrane mucin MUC1 can function as a physical barrier as well as a receptor for bacteria. MUC1 also influences epithelial cell morphology and receptor function. Various bacterial pathogens can exploit integrins to infect eukaryotic cells. It is yet unclear whether MUC1 influences the interaction of bacteria with integrins. We used Escherichia coli expressing the invasin (inv) protein of Yersinia pseudotuberculosis (E. coli inv) to assess the effects of MUC1 on ß1 integrin (ITGB1)-mediated bacterial invasion. Our results show that expression of full-length MUC1 does not yield a physical barrier but slightly enhances E. coli inv uptake. Enzymatic removal of the MUC1 extracellular domain (ED) using a secreted protease of C1 esterase inhibitor (StcE) of pathogenic Escherichia coli had no additional effect on E. coli inv invasion. In contrast, expression of a truncated MUC1 that lacks the cytoplasmic tail (CT) reduced bacterial entry substantially. Substitution of tyrosine residues in the MUC1 CT also reduced bacterial uptake, while deletion of the C-terminal half of the cytoplasmic tail only had a minor effect, pointing to a regulatory role of tyrosine phosphorylation and the N-terminal region of the MUC1 CT in integrin-mediated uptake process. Unexpectedly, StcE removal of the ED in MUC1-ΔCT cells reversed the block in bacterial invasion. Together, these findings indicate that MUC1 can facilitate ß1-integrin-mediated bacterial invasion by a concerted action of the large glycosylated extracellular domain and the membrane-juxtaposed cytoplasmic tail region.IMPORTANCE Bacteria can exploit membrane receptor integrins for cellular invasion, either by direct binding of bacterial adhesins or utilizing extracellular matrix components. MUC1 is a large transmembrane glycoprotein expressed by most epithelial cells that can have direct defensive or receptor functions at the host-microbe interface and is involved in facilitating integrin clustering. We investigated the role of epithelial MUC1 on ß1 integrin-mediated bacterial invasion. We discovered that MUC1 does not act as a barrier but facilitates bacterial entry through ß1 integrins. This process involves a concerted action of the MUC1 O-glycosylated extracellular domain and cytoplasmic tail. Our findings add a new dimension to the complexity of bacterial invasion mechanisms and provide novel insights into the distinct functions of MUC1 domains at the host-microbe interface.


Asunto(s)
Células Epiteliales/microbiología , Escherichia coli/metabolismo , Integrina beta1/metabolismo , Mucina-1/metabolismo , Yersinia pseudotuberculosis/genética , Adhesinas Bacterianas/genética , Escherichia coli/genética , Escherichia coli/patogenicidad , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Células HEK293 , Células HeLa , Humanos , Integrina beta1/genética , Mucina-1/genética
5.
PLoS Pathog ; 15(2): e1007566, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30716138

RESUMEN

The cellular invasion machinery of the enteric pathogen Salmonella consists of a type III secretion system (T3SS) with injectable virulence factors that induce uptake by macropinocytosis. Salmonella invasion at the apical surface of intestinal epithelial cells is inefficient, presumably because of a glycosylated barrier formed by transmembrane mucins that prevents T3SS contact with host cells. We observed that Salmonella is capable of apical invasion of intestinal epithelial cells that express the transmembrane mucin MUC1. Knockout of MUC1 in HT29-MTX cells or removal of MUC1 sialic acids by neuraminidase treatment reduced Salmonella apical invasion but did not affect lateral invasion that is not hampered by a defensive barrier. A Salmonella deletion strain lacking the SiiE giant adhesin was unable to invade intestinal epithelial cells through MUC1. SiiE-positive Salmonella closely associated with the MUC1 layer at the apical surface, but invaded Salmonella were negative for the adhesin. Our findings uncover that the transmembrane mucin MUC1 is required for Salmonella SiiE-mediated entry of enterocytes via the apical route.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Mucina-1/fisiología , Infecciones por Salmonella/metabolismo , Proteínas Bacterianas , Línea Celular , Elonguina/metabolismo , Enterocitos , Células Epiteliales , Humanos , Mucina-1/genética , Mucina-1/metabolismo , Salmonella enterica/patogenicidad , Salmonella typhimurium/patogenicidad , Factores de Virulencia
6.
Sci Rep ; 8(1): 10446, 2018 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-29993029

RESUMEN

P. aeruginosa is a notorious biofilm producer that causes a wide variety of acute and chronic infections. In this study the in vitro anti-biofilm activity of 13 Host Defence Peptides from different species was tested against P. aeruginosa biofilms. Most HDPs were able to prevent biofilm attachment, due to their antimicrobial effect on planktonic bacteria in the starting inoculum. Activity of HDPs against pre-formed biofilms was also observed, although mainly at short incubation times. Several HDPs were able to kill bacteria in the biofilm (colony counting of biofilm associated bacteria) but only CRAMP eradicated the whole biofilm (crystal violet staining). These results were quantitatively confirmed by confocal microscopy studies using a live/dead stain of the biofilms. Furthermore, for chicken CATH-2 (one of the more potent HDPs) it was shown that the peptide could indeed penetrate the biofilm structures and kill bacteria within the biofilm. These studies highlight the potency but also the limitations of HDPs as new potential anti-biofilm agents.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Biopelículas/efectos de los fármacos , Péptidos/farmacología , Pseudomonas aeruginosa/fisiología , Animales , Antibacterianos/farmacología , Catelicidinas/farmacología , Humanos , Pruebas de Sensibilidad Microbiana
7.
Cell Rep ; 10(4): 600-15, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25640182

RESUMEN

Itraconazole (ITZ) is a well-known antifungal agent that also has anticancer activity. In this study, we identify ITZ as a broad-spectrum inhibitor of enteroviruses (e.g., poliovirus, coxsackievirus, enterovirus-71, rhinovirus). We demonstrate that ITZ inhibits viral RNA replication by targeting oxysterol-binding protein (OSBP) and OSBP-related protein 4 (ORP4). Consistently, OSW-1, a specific OSBP/ORP4 antagonist, also inhibits enterovirus replication. Knockdown of OSBP inhibits virus replication, whereas overexpression of OSBP or ORP4 counteracts the antiviral effects of ITZ and OSW-1. ITZ binds OSBP and inhibits its function, i.e., shuttling of cholesterol and phosphatidylinositol-4-phosphate between membranes, thereby likely perturbing the virus-induced membrane alterations essential for viral replication organelle formation. ITZ also inhibits hepatitis C virus replication, which also relies on OSBP. Together, these data implicate OSBP/ORP4 as molecular targets of ITZ and point to an essential role of OSBP/ORP4-mediated lipid exchange in virus replication that can be targeted by antiviral drugs.


Asunto(s)
Enterovirus/efectos de los fármacos , Enterovirus/metabolismo , Itraconazol/farmacología , Receptores de Esteroides/metabolismo , Replicación Viral/efectos de los fármacos , Antivirales/farmacología , Línea Celular Tumoral , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA