Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
EMBO J ; 41(19): e110629, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-35968812

RESUMEN

Dysbiosis of vaginal microbiota is associated with increased HIV-1 acquisition, but the underlying cellular mechanisms remain unclear. Vaginal Langerhans cells (LCs) protect against mucosal HIV-1 infection via autophagy-mediated degradation of HIV-1. As LCs are in continuous contact with bacterial members of the vaginal microbiome, we investigated the impact of commensal and dysbiosis-associated vaginal (an)aerobic bacterial species on the antiviral function of LCs. Most of the tested bacteria did not affect the HIV-1 restrictive function of LCs. However, Prevotella timonensis induced a vast uptake of HIV-1 by vaginal LCs. Internalized virus remained infectious for days and uptake was unaffected by antiretroviral drugs. P. timonensis-exposed LCs efficiently transmitted HIV-1 to target cells both in vitro and ex vivo. Additionally, P. timonensis exposure enhanced uptake and transmission of the HIV-1 variants that establish infection after sexual transmission, the so-called Transmitted Founder variants. Our findings, therefore, suggest that P. timonensis might set the stage for enhanced HIV-1 susceptibility during vaginal dysbiosis and advocate targeted treatment of P. timonensis during bacterial vaginosis to limit HIV-1 infection.


Asunto(s)
Infecciones por VIH , VIH-1 , Antivirales , Disbiosis , Femenino , Humanos , Células de Langerhans , Prevotella
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.
PLoS Pathog ; 19(8): e1011571, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37561789

RESUMEN

Mucins play an essential role in protecting the respiratory tract against microbial infections while also acting as binding sites for bacterial and viral adhesins. The heavily O-glycosylated gel-forming mucins MUC5AC and MUC5B eliminate pathogens by mucociliary clearance. Transmembrane mucins MUC1, MUC4, and MUC16 can restrict microbial invasion at the apical surface of the epithelium. In this study, we determined the impact of host mucins and mucin glycans on epithelial entry of SARS-CoV-2. Human lung epithelial Calu-3 cells express the SARS-CoV-2 entry receptor ACE2 and high levels of glycosylated MUC1, but not MUC4 and MUC16, on their cell surface. The O-glycan-specific mucinase StcE specifically removed the glycosylated part of the MUC1 extracellular domain while leaving the underlying SEA domain and cytoplasmic tail intact. StcE treatment of Calu-3 cells significantly enhanced infection with SARS-CoV-2 pseudovirus and authentic virus, while removal of terminal mucin glycans sialic acid and fucose from the epithelial surface did not impact viral entry. In Calu-3 cells, the transmembrane mucin MUC1 and ACE2 are located to the apical surface in close proximity and StcE treatment results in enhanced binding of purified spike protein. Both MUC1 and MUC16 are expressed on the surface of human organoid-derived air-liquid interface (ALI) differentiated airway cultures and StcE treatment led to mucin removal and increased levels of SARS-CoV-2 replication. In these cultures, MUC1 was highly expressed in non-ciliated cells while MUC16 was enriched in goblet cells. In conclusion, the glycosylated extracellular domains of different transmembrane mucins might have similar protective functions in different respiratory cell types by restricting SARS-CoV-2 binding and entry.


Asunto(s)
COVID-19 , Mucinas , Humanos , Mucinas/metabolismo , Enzima Convertidora de Angiotensina 2 , SARS-CoV-2/metabolismo , Antígeno Ca-125/metabolismo , Pulmón/metabolismo , Polisacáridos
4.
J Infect Dis ; 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38573164

RESUMEN

Dysbiosis of the vaginal microbiome poses a serious risk for sexual HIV-1 transmission. Prevotella spp. are abundant during vaginal dysbiosis and associated with enhanced HIV-1 susceptibility; however, underlying mechanisms remain unclear. Here, we investigated the direct effect of vaginal bacteria on HIV-1 susceptibility of vaginal CD4+ T cells. Notably, pre-exposure to Prevotella timonensis enhanced HIV-1 uptake by vaginal T cells, leading to increased viral fusion and enhanced virus production. Pre-exposure to antiretroviral inhibitors abolished Prevotella timonensis-enhanced infection. Hence, our study shows that the vaginal microbiome directly affects mucosal CD4+ T cell susceptibility, emphasising importance of vaginal dysbiosis diagnosis and treatment.

5.
Molecules ; 27(5)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35268716

RESUMEN

Fucosidases are associated with several pathological conditions and play an important role in the health of the human gut. For example, fucosidases have been shown to be indicators and/or involved in hepatocellular carcinoma, breast cancer, and helicobacter pylori infections. A prerequisite for the detection and profiling of fucosidases is the formation of a specific covalent linkage between the enzyme of interest and the activity-based probe (ABP). The most commonly used fucosidase ABPs are limited to only one of the classes of fucosidases, the retaining fucosidases. New approaches are needed that allow for the detection of the second class of fucosidases, the inverting type. Here, we report an ortho-quinone methide-based probe with an azide mini-tag that selectively labels both retaining and inverting bacterial α-l-fucosidases. Mass spectrometry-based intact protein and sequence analysis of a probe-labeled bacterial fucosidase revealed almost exclusive single labeling at two specific tryptophan residues outside of the active site. Furthermore, the probe could detect and image extracellular fucosidase activity on the surface of live bacteria.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Indolquinonas , Helicobacter pylori/metabolismo , Humanos , alfa-L-Fucosidasa/metabolismo
6.
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
7.
Cell Microbiol ; 22(12): e13252, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32827216

RESUMEN

The enteropathogenic bacterium, Campylobacter jejuni, was considered to be non-saccharolytic, but recently it emerged that l-fucose plays a central role in C. jejuni virulence. Half of C. jejuni clinical isolates possess an operon for l-fucose utilisation. In the intestinal tract, l-fucose is abundantly available in mucin O-linked glycan structures, but C. jejuni lacks a fucosidase enzyme essential to release the l-fucose. We set out to determine how C. jejuni can gain access to these intestinal l-fucosides. Growth of the fuc + C. jejuni strains, 129,108 and NCTC 11168, increased in the presence of l-fucose while fucose permease knockout strains did not benefit from additional l-fucose. With fucosidase assays and an activity-based probe, we confirmed that Bacteriodes fragilis, an abundant member of the intestinal microbiota, secretes active fucosidases. In the presence of mucins, C. jejuni was dependent on B. fragilis fucosidase activity for increased growth. Campylobacter jejuni invaded Caco-2 intestinal cells that express complex O-linked glycan structures that contain l-fucose. In infection experiments, C. jejuni was more invasive in the presence of B. fragilis and this increase is due to fucosidase activity. We conclude that C. jejuni fuc + strains are dependent on exogenous fucosidases for increased growth and invasion.


Asunto(s)
Bacteroides fragilis/enzimología , Campylobacter jejuni/crecimiento & desarrollo , Campylobacter jejuni/patogenicidad , Fucosa/metabolismo , Mucinas/metabolismo , alfa-L-Fucosidasa/metabolismo , Células CACO-2 , Campylobacter jejuni/genética , Humanos , Interacciones Microbianas/fisiología , Virulencia , alfa-L-Fucosidasa/biosíntesis
8.
Org Biomol Chem ; 19(13): 2968-2977, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33729259

RESUMEN

GH29 α-l-fucosidases catalyze hydrolysis of terminal α-l-fucosyl linkages with varying specificity and are expressed by prominent members of the human gut microbiota. Both homeostasis and dysbiosis at the human intestinal microbiota interface have been correlated with altered fucosidase activity. Herein we describe the development of a 2-deoxy-2-fluoro fucosyl fluoride derivative with an azide mini-tag as an activity-based probe (ABP) for selective in vitro labelling of GH29 α-l-fucosidases. Only catalytically active fucosidases are inactivated by this ABP, allowing their functionalization with a biotin reporter group via the CuAAC reaction and subsequent in-gel detection at nanogram levels. The ABP we present here is shown to be active against a GH29 α-l-fucosidase from Bacteroides fragilis and capable of labeling two other GH29 α-l-fucosidases with different linkage specificity, illustrating its broader utility. This novel ABP is a valuable addition to the toolbox of fucosidase probes by allowing identification and functional studies of the wide variety of GH29 fucosidases, including those in the gut microbiota.


Asunto(s)
Fucosa/química , Sondas Moleculares/química , alfa-L-Fucosidasa/análisis , Bacteroides fragilis/enzimología , Fucosa/análogos & derivados , Fucosa/farmacología , Microbioma Gastrointestinal , Humanos , Sondas Moleculares/síntesis química , Sondas Moleculares/farmacología , Estructura Molecular , alfa-L-Fucosidasa/antagonistas & inhibidores , alfa-L-Fucosidasa/metabolismo
9.
Cell Microbiol ; 19(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28008697

RESUMEN

Flagella are nanofibers that drive bacterial movement. The filaments are generally composed of thousands of tightly packed flagellin subunits with a terminal cap protein, named FliD. Here, we report that the FliD protein of the bacterial pathogen Campylobacter jejuni binds to host cells. Live-cell imaging and confocal microscopy showed initial contact of the bacteria with epithelial cells via the flagella tip. Recombinant FliD protein bound to the surface of intestinal epithelial cells in a dose-dependent fashion. Search for the FliD binding site on the host cell using cells with defined glycosylation defects indicated glycosaminoglycans as a putative target. Heparinase treatment of wild type cells and an excess of soluble heparin abolished FliD binding. Binding assays showed direct and specific binding of FliD to heparin. Addition of an excess of purified FliD or heparin reduced the attachment of viable C. jejuni to the host cells. The host cell binding domain of FliD was mapped to the central region of the protein. Overall, our results indicate that the C. jejuni flagellar tip protein FliD acts as an attachment factor that interacts with cell surface heparan sulfate glycosaminoglycan receptors.


Asunto(s)
Adhesión Bacteriana/fisiología , Proteínas Bacterianas/metabolismo , Campylobacter jejuni/metabolismo , Flagelos/metabolismo , Glicosaminoglicanos/metabolismo , Mucosa Intestinal/parasitología , Animales , Adhesión Bacteriana/efectos de los fármacos , Proteínas Bacterianas/genética , Sitios de Unión/fisiología , Células CHO , Infecciones por Campylobacter/microbiología , Infecciones por Campylobacter/patología , Línea Celular Tumoral , Cricetulus , Células Epiteliales/citología , Células Epiteliales/parasitología , Flagelina/metabolismo , Células HT29 , Liasa de Heparina/farmacología , Humanos , Mucosa Intestinal/citología
10.
PLoS Pathog ; 11(10): e1005188, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26431038

RESUMEN

The ability of phagocytes to clear pathogens is an essential attribute of the innate immune response. The role of signaling lipid molecules such as phosphoinositides is well established, but the role of membrane sphingolipids in phagocytosis is largely unknown. Using a genetic approach and small molecule inhibitors, we show that phagocytosis of Candida albicans requires an intact sphingolipid biosynthetic pathway. Blockade of serine-palmitoyltransferase (SPT) and ceramide synthase-enzymes involved in sphingolipid biosynthesis- by myriocin and fumonisin B1, respectively, impaired phagocytosis by phagocytes. We used CRISPR/Cas9-mediated genome editing to generate Sptlc2-deficient DC2.4 dendritic cells, which lack serine palmitoyl transferase activity. Sptlc2-/- DC2.4 cells exhibited a stark defect in phagocytosis, were unable to bind fungal particles and failed to form a normal phagocytic cup to engulf C. albicans. Supplementing the growth media with GM1, the major ganglioside present at the cell surface, restored phagocytic activity of Sptlc2-/- DC2.4 cells. While overall membrane trafficking and endocytic pathways remained functional, Sptlc2-/- DC2.4 cells express reduced levels of the pattern recognition receptors Dectin-1 and TLR2 at the cell surface. Consistent with the in vitro data, compromised sphingolipid biosynthesis in mice sensitizes the animal to C. albicans infection. Sphingolipid biosynthesis is therefore critical for phagocytosis and in vivo clearance of C. albicans.


Asunto(s)
Candida albicans , Candidiasis/inmunología , Interacciones Huésped-Patógeno/inmunología , Fagocitosis/fisiología , Esfingolípidos/biosíntesis , Animales , Candida albicans/inmunología , Línea Celular , Cromatografía en Capa Delgada , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Modelos Animales de Enfermedad , Citometría de Flujo , Técnicas de Inactivación de Genes , Humanos , Espectrometría de Masas , Ratones
11.
Eukaryot Cell ; 14(2): 182-93, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25548150

RESUMEN

Membrane reshaping resides at the core of many important cellular processes, and among its mediators are the BAR (Bin, Amphiphysin, Rvs) domain-containing proteins. We have explored the diversity and function of the Rvs BAR proteins in Candida albicans and identified a novel family member, Rvs167-3 (orf19.1861). We show that Rvs167-3 specifically interacts with Rvs162 to form a stable BAR heterodimer able to bind liposomes in vitro. A second, distinct heterodimer is formed by the canonical BAR proteins Rvs161 and Rvs167. Purified Rvs161/Rvs167 complex also binds liposomes, indicating that C. albicans expresses two functional BAR heterodimers. We used live-cell imaging to localize green fluorescent protein (GFP)-tagged Rvs167-3 and Rvs167 and show that both proteins concentrate in small cortical spots. However, while Rvs167 strictly colocalizes with the endocytic marker protein Abp1, we do not observe any colocalization of Rvs167-3 with sites of endocytosis marked by Abp1. Furthermore, the rvs167-3Δ/Δ mutant is not defective in endocytosis and strains lacking Rvs167-3 or its partner Rvs162 do not display increased sensitivity to high salt concentrations or decreased cell wall integrity, phenotypes which have been observed for rvs167Δ/Δ and rvs161Δ/Δ strains and which are linked to endocytosis defects. Taken together, our results indicate different roles for the two BAR heterodimers in C. albicans: the canonical Rvs161/Rvs167 heterodimer functions in endocytosis, whereas the novel Rvs162/Rvs167-3 heterodimer seems not to be involved in this process. Nevertheless, despite their different roles, our phenotypic analysis revealed a genetic interaction between the two BAR heterodimers, suggesting that they may have related but distinct membrane-associated functions.


Asunto(s)
Candida albicans/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas Fúngicas/metabolismo , Candida albicans/metabolismo , Membrana Celular/metabolismo , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Endocitosis , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas
12.
PLoS Pathog ; 9(6): e1003446, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825946

RESUMEN

Phagocytosis of the opportunistic fungal pathogen Candida albicans by cells of the innate immune system is vital to prevent infection. Dectin-1 is the major phagocytic receptor involved in anti-fungal immunity. We identify two new interacting proteins of Dectin-1 in macrophages, Bruton's Tyrosine Kinase (BTK) and Vav1. BTK and Vav1 are recruited to phagocytic cups containing C. albicans yeasts or hyphae but are absent from mature phagosomes. BTK and Vav1 localize to cuff regions surrounding the hyphae, while Dectin-1 lines the full length of the phagosome. BTK and Vav1 colocalize with the lipid PI(3,4,5)P3 and F-actin at the phagocytic cup, but not with diacylglycerol (DAG) which marks more mature phagosomal membranes. Using a selective BTK inhibitor, we show that BTK contributes to DAG synthesis at the phagocytic cup and the subsequent recruitment of PKCε. BTK- or Vav1-deficient peritoneal macrophages display a defect in both zymosan and C. albicans phagocytosis. Bone marrow-derived macrophages that lack BTK or Vav1 show reduced uptake of C. albicans, comparable to Dectin1-deficient cells. BTK- or Vav1-deficient mice are more susceptible to systemic C. albicans infection than wild type mice. This work identifies an important role for BTK and Vav1 in immune responses against C. albicans.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Proteínas de Homeodominio/inmunología , Lectinas Tipo C/inmunología , Macrófagos Peritoneales/inmunología , Neuropéptidos/inmunología , Fagocitosis/inmunología , Proteínas Tirosina Quinasas/inmunología , Actinas/genética , Actinas/inmunología , Actinas/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Candida albicans/metabolismo , Candidiasis/genética , Candidiasis/metabolismo , Candidiasis/patología , Línea Celular , Diglicéridos/genética , Diglicéridos/inmunología , Diglicéridos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Noqueados , Neuropéptidos/genética , Neuropéptidos/metabolismo , Fagocitosis/genética , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/inmunología , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo
13.
Traffic ; 13(6): 780-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22348280

RESUMEN

Sortagging is a versatile method for site-specific modification of proteins as applied to a variety of in vitro reactions. Here, we explore possibilities of adapting the sortase method for use in living cells. For intracellular sortagging, we employ the Ca²âº-independent sortase A transpeptidase (SrtA) from Streptococcus pyogenes. Substrate proteins were equipped with the C-terminal sortase-recognition motif (LPXTG); we used proteins with an N-terminal (oligo)glycine as nucleophiles. We show that sortase-dependent protein ligation can be achieved in Saccharomyces cerevisiae and in mammalian HEK293T cells, both in the cytosol and in the lumen of the endoplasmic reticulum (ER). ER luminal sortagging enables secretion of the reaction products, among which circular polypeptides. Protein ligation of substrate and nucleophile occurs within 30 min of translation. The versatility of the method is shown by protein ligation of multiple substrates with green fluorescent protein-based nucleophiles in different intracellular compartments.


Asunto(s)
Aminoaciltransferasas/fisiología , Proteínas Bacterianas/fisiología , Cisteína Endopeptidasas/fisiología , Streptococcus pyogenes/metabolismo , Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Biología Celular , Cisteína Endopeptidasas/metabolismo , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Cinética , Espectrometría de Masas/métodos , Péptidos/química , Plásmidos/metabolismo , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/metabolismo
14.
Proc Natl Acad Sci U S A ; 108(34): 14270-5, 2011 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-21825168

RESUMEN

Dectin-1, the major ß-glucan receptor in leukocytes, triggers an effective immune response upon fungal recognition. Here we use sortase-mediated transpeptidation, a technique that allows placement of a variety of probes on a polypeptide backbone, to monitor the behavior of labeled functional dectin-1 in live cells with and without fungal challenge. Installation of probes on dectin-1 by sortagging permitted highly specific visualization of functional protein on the cell surface and its subsequent internalization upon ligand presentation. Retrieval of sortagged dectin-1 expressed in macrophages uncovered a unique interaction between dectin-1 and galectin-3 that functions in the proinflammatory response of macrophages to pathogenic fungi. When macrophages expressing dectin-1 are exposed to Candida albicans mutants with increased exposure of ß-glucan, the loss of galectin-3 dramatically accentuates the failure to trigger an appropriate TNF-α response.


Asunto(s)
Candida albicans/fisiología , Galectina 3/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Saccharomyces cerevisiae/fisiología , Animales , Biotinilación , Células de la Médula Ósea/citología , Endocitosis/efectos de los fármacos , Células HEK293 , Humanos , Inmunoprecipitación , Lectinas Tipo C , Ratones , Sondas Moleculares/metabolismo , Unión Proteica , Coloración y Etiquetado , Factor de Necrosis Tumoral alfa/metabolismo , Zimosan/metabolismo , beta-Glucanos/metabolismo
15.
Life Sci Alliance ; 7(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38508723

RESUMEN

Mucin 1 (MUC1) is a transmembrane mucin expressed at the apical surface of epithelial cells at mucosal surfaces. MUC1 has a barrier function against bacterial invasion and is well known for its aberrant expression and glycosylation in adenocarcinomas. The MUC1 extracellular domain contains a variable number of tandem repeats (VNTR) of 20 amino acids, which are heavily O-linked glycosylated. Monoclonal antibodies against the MUC1 VNTR are powerful research tools with applications in the diagnosis and treatment of MUC1-expressing cancers. Here, we report direct mass spectrometry-based sequencing of anti-MUC1 hybridoma-derived 139H2 IgG, enabling reverse-engineering of the functional recombinant monoclonal antibody. The crystal structure of the 139H2 Fab fragment in complex with the MUC1 epitope was solved, revealing the molecular basis of 139H2 binding specificity to MUC1 and its tolerance to O-glycosylation of the VNTR. The available sequence of 139H2 will allow further development of MUC1-related diagnostic, targeting, and treatment strategies.


Asunto(s)
Mucina-1 , Neoplasias , Humanos , Secuencia de Aminoácidos , Mucina-1/genética , Mucina-1/química , Mucinas/genética , Mucinas/metabolismo , Glicosilación , Anticuerpos Monoclonales
16.
Toxins (Basel) ; 15(10)2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37888617

RESUMEN

Clostridioides difficile infection is expected to become the most common healthcare-associated infection worldwide. C. difficile-induced pathogenicity is significantly attributed to its enterotoxin, TcdA, which primarily targets Rho-GTPases involved in regulating cytoskeletal and tight junction (TJ) dynamics, thus leading to cytoskeleton breakdown and ultimately increased intestinal permeability. This study investigated whether two non-digestible oligosaccharides (NDOs), alginate (AOS) and chitosan (COS) oligosaccharides, possess antipathogenic and barrier-protective properties against C. difficile bacteria and TcdA toxin, respectively. Both NDOs significantly reduced C. difficile growth, while cell cytotoxicity assays demonstrated that neither COS nor AOS significantly attenuated the TcdA-induced cell death 24 h post-exposure. The challenge of Caco-2 monolayers with increasing TcdA concentrations increased paracellular permeability, as measured by TEER and LY flux assays. In this experimental setup, COS completely abolished, and AOS mitigated, the deleterious effects of TcdA on the monolayer's integrity. These events were not accompanied by alterations in ZO-1 and occludin protein levels; however, immunofluorescence microscopy revealed that both AOS and COS prevented the TcdA-induced occludin mislocalization. Finally, both NDOs accelerated TJ reassembly upon a calcium-switch assay. Overall, this study established the antipathogenic and barrier-protective capacity of AOS and COS against C. difficile and its toxin, TcdA, while revealing their ability to promote TJ reassembly in Caco-2 cells.


Asunto(s)
Toxinas Bacterianas , Quitosano , Clostridioides difficile , Humanos , Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Células CACO-2 , Quitosano/farmacología , Clostridioides/metabolismo , Alginatos/farmacología , Ocludina , Enterotoxinas/toxicidad , Enterotoxinas/metabolismo , Oligosacáridos/farmacología , Oligosacáridos/metabolismo
17.
FEMS Yeast Res ; 12(1): 61-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22094058

RESUMEN

The pentose phosphate pathway (PPP) is the main source of NADPH in the cell and therefore essential for the maintenance of the redox balance and anabolic reactions. NADPH is produced by the two dehydrogenases in the oxidative branch of the PPP: glucose-6-phosphate dehydrogenase (Zwf1) and 6-phosphogluconate dehydrogenase (Gnd1). We observed that in the commensal fungus Candida albicans these two enzymes contain putative peroxisomal targeting signals (PTSs): Zwf1 has a putative PTS1, while the annotated intron of GND1 encodes a PTS2. By subcellular fractionation and fluorescence microscopy, we show that both enzymes have a dual localization in which the majority is cytosolic, but a small fraction is peroxisome associated. Analysis of GND1 transcripts revealed that dual targeting of Gnd1 is directed by alternative splicing resulting in two Gnd1 isoforms, one without targeting signals localized to the cytosol and one with an N-terminal PTS2 targeted to peroxisomes. To our knowledge, Gnd1 is the first example of dual targeting of a protein by alternative splicing in C. albicans. In silico analysis suggests that PTS-mediated peroxisomal targeting of Zwf1 and Gnd1 is conserved across closely related Candida species. We discuss putative functions of the peroxisomal oxidative PPP in these organisms.


Asunto(s)
Empalme Alternativo , Candida albicans/enzimología , Candida albicans/genética , Citosol/enzimología , Peroxisomas/enzimología , Fosfogluconato Deshidrogenasa/genética , Fosfogluconato Deshidrogenasa/metabolismo , Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/metabolismo , Microscopía Fluorescente , Señales de Clasificación de Proteína
18.
J Biol Chem ; 285(32): 24335-46, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20522553

RESUMEN

Transport of acetyl-CoA between intracellular compartments is mediated by carnitine acetyltransferases (Cats) that reversibly link acetyl units to the carrier molecule carnitine. The genome of the opportunistic pathogenic yeast Candida albicans encodes several (putative) Cats: the peroxisomal and mitochondrial Cat2 isoenzymes encoded by a single gene and the carnitine acetyltransferase homologs Yat1 and Yat2. To determine the contributions of the individual Cats, various carnitine acetyltransferase mutant strains were constructed and subjected to phenotypic and biochemical analyses on different carbon sources. We show that mitochondrial Cat2 is required for the intramitochondrial conversion of acetylcarnitine to acetyl-CoA, which is essential for a functional tricarboxylic acid cycle during growth on oleate, acetate, ethanol, and citrate. Yat1 is cytosolic and contributes to acetyl-CoA transport from the cytosol during growth on ethanol or acetate, but its activity is not required for growth on oleate. Yat2 is also cytosolic, but we were unable to attribute any function to this enzyme. Surprisingly, peroxisomal Cat2 is essential neither for export of acetyl units during growth on oleate nor for the import of acetyl units during growth on acetate or ethanol. Oxidation of fatty acids still takes place in the absence of peroxisomal Cat2, but biomass formation is absent, and the strain displays a growth delay on acetate and ethanol that can be partially rescued by the addition of carnitine. Based on our results, we present a model for the intracellular flow of acetyl units under various growth conditions and the roles of each of the Cats in this process.


Asunto(s)
Candida albicans/enzimología , Carnitina O-Acetiltransferasa/metabolismo , Transporte Biológico , Carbono/química , Carnitina O-Acetiltransferasa/química , Membrana Celular/metabolismo , Ácidos Grasos/química , Espectrometría de Masas/métodos , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Modelos Biológicos , Mutación , Oxígeno/química , Peroxisomas/química , Peroxisomas/metabolismo , Fenotipo , Proteínas de Saccharomyces cerevisiae/metabolismo , Técnicas del Sistema de Dos Híbridos
19.
Eukaryot Cell ; 9(12): 1809-15, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20889721

RESUMEN

Acetyl coenzyme A (acetyl-CoA) is a central metabolite in carbon and energy metabolism. Because of its amphiphilic nature and bulkiness, acetyl-CoA cannot readily traverse biological membranes. In fungi, two systems for acetyl unit transport have been identified: a shuttle dependent on the carrier carnitine and a (peroxisomal) citrate synthase-dependent pathway. In the carnitine-dependent pathway, carnitine acetyltransferases exchange the CoA group of acetyl-CoA for carnitine, thereby forming acetyl-carnitine, which can be transported between subcellular compartments. Citrate synthase catalyzes the condensation of oxaloacetate and acetyl-CoA to form citrate that can be transported over the membrane. Since essential metabolic pathways such as fatty acid ß-oxidation, the tricarboxylic acid (TCA) cycle, and the glyoxylate cycle are physically separated into different organelles, shuttling of acetyl units is essential for growth of fungal species on various carbon sources such as fatty acids, ethanol, acetate, or citrate. In this review we summarize the current knowledge on the different systems of acetyl transport that are operational during alternative carbon metabolism, with special focus on two fungal species: Saccharomyces cerevisiae and Candida albicans.


Asunto(s)
Acetiltransferasas/metabolismo , Candida albicans/enzimología , Carbono/metabolismo , Proteínas Fúngicas/metabolismo , Saccharomyces cerevisiae/enzimología , Acetilcoenzima A/metabolismo , Acetiltransferasas/genética , Candida albicans/genética , Candida albicans/metabolismo , Proteínas Fúngicas/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA