Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 134(9)2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38483511

RESUMEN

In lung, thromboxane A2 (TXA2) activates the TP receptor to induce proinflammatory and bronchoconstrictor effects. Thus, TP receptor antagonists and TXA2 synthase inhibitors have been tested as potential asthma therapeutics in humans. Th9 cells play key roles in asthma and regulate the lung immune response to allergens. Herein, we found that TXA2 reduces Th9 cell differentiation during allergic lung inflammation. Th9 cells were decreased approximately 2-fold and airway hyperresponsiveness was attenuated in lungs of allergic mice treated with TXA2. Naive CD4+ T cell differentiation to Th9 cells and IL-9 production were inhibited dose-dependently by TXA2 in vitro. TP receptor-deficient mice had an approximately 2-fold increase in numbers of Th9 cells in lungs in vivo after OVA exposure compared with wild-type mice. Naive CD4+ T cells from TP-deficient mice exhibited increased Th9 cell differentiation and IL-9 production in vitro compared with CD4+ T cells from wild-type mice. TXA2 also suppressed Th2 and enhanced Treg differentiation both in vitro and in vivo. Thus, in contrast to its acute, proinflammatory effects, TXA2 also has longer-lasting immunosuppressive effects that attenuate the Th9 differentiation that drives asthma progression. These findings may explain the paradoxical failure of anti-thromboxane therapies in the treatment of asthma.


Asunto(s)
Asma , Diferenciación Celular , Linfocitos T Reguladores , Células Th2 , Tromboxano A2 , Animales , Ratones , Células Th2/inmunología , Células Th2/patología , Tromboxano A2/metabolismo , Tromboxano A2/inmunología , Linfocitos T Reguladores/inmunología , Asma/inmunología , Asma/patología , Asma/tratamiento farmacológico , Asma/genética , Ratones Noqueados , Interleucina-9/inmunología , Interleucina-9/genética , Interleucina-9/metabolismo , Neumonía/inmunología , Neumonía/patología , Ratones Endogámicos C57BL , Ratones Endogámicos BALB C , Pulmón/inmunología , Pulmón/patología , Ovalbúmina/inmunología , Femenino , Linfocitos T Colaboradores-Inductores/inmunología
2.
Mol Immunol ; 156: 127-135, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36921487

RESUMEN

The lung airway epithelial surface is heavily covered with sialic acids as the terminal carbohydrate on most cell surface glycoconjugates and can be removed by microbial neuraminidases or endogenous sialidases. By desialylating the lung epithelial surface, neuraminidase acts as an important virulence factor in many mucosal pathogens, such as influenza and S. pneumoniae. Desialylation exposes the subterminal galactosyl moieties - the binding glycotopes for galectins, a family of carbohydrate-recognition proteins playing important roles in various aspects of immune responses. Galectin-1 and galectin-3 have been extensively studied in their roles related to host immune responses, but some questions about their role(s) in leukocyte recruitment during lung bacterial infection remain unanswered. In this study, we found that both galectin-1 and galectin-3 bind to polymorphonuclear leukocytes (PMNs) and enhance the interaction of endothelial intercellular adhesion molecule-1 (ICAM-1) with PMNs, which is further increased by PMN desialylation. In addition, we observed that in vitro galectin-1 mediates the binding of PMNs, particularly desialylated PMNs, onto the endothelial cells. Finally, in a murine model for LPS-mediated acute lung injury, we observed that galectin-1 modulates PMN infiltration to the lung without altering the expression of chemoattractant cytokines. We conclude that galectins, particularly galectin-1, may function as adhesion molecules that mediate PMN-endothelial cell interactions, and modulate PMN infiltration during acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda , Neutrófilos , Humanos , Ratones , Animales , Lipopolisacáridos/farmacología , Células Endoteliales , Galectina 1 , Galectina 3 , Adhesión Celular , Pulmón , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Streptococcus pneumoniae , Molécula 1 de Adhesión Intercelular/metabolismo
3.
Bioorg Med Chem ; 72: 116974, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36108470

RESUMEN

Human galectin 3 (Gal-3) has been implicated to play important roles in different biological recognition processes such as tumor growth and cancer metastasis. High-affinity Gal-3 ligands are desirable for functional studies and as inhibitors for potential therapeutic development. We report here a facile synthesis of ß-cyclodextrin (CD)-based Tn and TF antigen-containing multivalent ligands via a click reaction. Binding studies indicated that the synthetic multivalent glycan ligands demonstrated a clear clustering effect in binding to human Gal-3, with up to 153-fold enhanced relative affinity in comparison with the monomeric glycan ligand. The GalNAc (Tn antigen) containing heptavalent ligand showed the highest affinity for human Gal-3 among the synthetic ligands tested, with an EC50 of 1.4 µM in binding to human Gal-3. A cell-based assay revealed that the synthetic CD-based multivalent ligands could efficiently inhibit Gal-3 binding to human airway epithelial cells, with an inhibitory capacity consistent with their binding affinity measured by SPR. The synthetic cyclodextrin-based ligands described in this study should be valuable for functional studies of human Gal-3 and potentially for therapeutic applications.


Asunto(s)
Ciclodextrinas , beta-Ciclodextrinas , Galectina 3/metabolismo , Humanos , Ligandos , Unión Proteica , beta-Ciclodextrinas/farmacología
4.
Sci Rep ; 12(1): 14173, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35986080

RESUMEN

To gain insight into sialic acid biology and sialidase/neuraminidase (NEU) expression in mature human neutrophil (PMN)s, we studied NEU activity and expression in PMNs and the HL60 promyelocytic leukemic cell line, and changes that might occur in PMNs undergoing apoptosis and HL60 cells during their differentiation into PMN-like cells. Mature human PMNs contained NEU activity and expressed NEU2, but not NEU1, the NEU1 chaperone, protective protein/cathepsin A(PPCA), NEU3, and NEU4 proteins. In proapoptotic PMNs, NEU2 protein expression increased > 30.0-fold. Granulocyte colony-stimulating factor protected against NEU2 protein upregulation, PMN surface desialylation and apoptosis. In response to 3 distinct differentiating agents, dimethylformamide, dimethylsulfoxide, and retinoic acid, total NEU activity in differentiated HL60 (dHL60) cells was dramatically reduced compared to that of nondifferentiated cells. With differentiation, NEU1 protein levels decreased > 85%, PPCA and NEU2 proteins increased > 12.0-fold, and 3.0-fold, respectively, NEU3 remained unchanged, and NEU4 increased 1.7-fold by day 3, and then returned to baseline. In dHL60 cells, lectin blotting revealed decreased α2,3-linked and increased α2,6-linked sialylation. dHL60 cells displayed increased adhesion to and migration across human bone marrow-derived endothelium and increased bacterial phagocytosis. Therefore, myeloid apoptosis and differentiation provoke changes in NEU catalytic activity and protein expression, surface sialylation, and functional responsiveness.


Asunto(s)
Ácido N-Acetilneuramínico , Neuraminidasa , Apoptosis , Diferenciación Celular , Humanos , Ácido N-Acetilneuramínico/metabolismo , Neuraminidasa/metabolismo , Neutrófilos/metabolismo
5.
Methods Mol Biol ; 2442: 425-443, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35320539

RESUMEN

Techniques for disrupting gene expression are invaluable tools for the analysis of the biological role of a gene product. Because of its genetic tractability and multiple advantages over conventional mammalian models, the zebrafish (Danio rerio) is recognized as a powerful system for gaining new insight into diverse aspects of human health and disease. Among the multiple mammalian gene families for which the zebrafish has shown promise as an invaluable model for functional studies, the galectins have attracted great interest due to their participation in early development, regulation of immune homeostasis, and recognition of microbial pathogens. Galectins are ß-galactosyl-binding lectins with a characteristic sequence motif in their carbohydrate recognition domains (CRDs), that constitute an evolutionary conserved family ubiquitous in eukaryotic taxa. Galectins are emerging as key players in the modulation of many important pathological processes, which include acute and chronic inflammatory diseases, autoimmunity and cancer, thus making them potential molecular targets for innovative drug discovery. Here, we provide a review of the current methods available for the manipulation of gene expression in the zebrafish, with a focus on gene knockdown [morpholino (MO)-derived antisense oligonucleotides] and knockout (CRISPR-Cas) technologies.


Asunto(s)
Galectinas , Pez Cebra , Animales , Galectinas/metabolismo , Técnicas de Silenciamiento del Gen , Mamíferos/genética , Morfolinos/genética , Morfolinos/metabolismo , ARN/metabolismo , Pez Cebra/metabolismo
6.
J Exp Med ; 218(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33216117

RESUMEN

Two cosegregating single-nucleotide polymorphisms (SNPs) in human TLR4, an A896G transition at SNP rs4986790 (D299G) and a C1196T transition at SNP rs4986791 (T399I), have been associated with LPS hyporesponsiveness and differential susceptibility to many infectious or inflammatory diseases. However, many studies failed to confirm these associations, and transfection experiments resulted in conflicting conclusions about the impact of these SNPs on TLR4 signaling. Using advanced protein modeling from crystallographic data of human and murine TLR4, we identified homologous substitutions of these SNPs in murine Tlr4, engineered a knock-in strain expressing the D298G and N397I TLR4 SNPs homozygously, and characterized in vivo and in vitro responses to TLR4 ligands and infections in which TLR4 is implicated. Our data provide new insights into cellular and molecular mechanisms by which these SNPs decrease the TLR4 signaling efficiency and offer an experimental approach to confirm or refute human data possibly confounded by variables unrelated to the direct effects of the SNPs on TLR4 functionality.


Asunto(s)
Lipopolisacáridos/genética , Polimorfismo de Nucleótido Simple/genética , Receptor Toll-Like 4/genética , Animales , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Ratones , Transducción de Señal/genética
7.
Glycobiology ; 30(11): 895-909, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-32280962

RESUMEN

Pregnancy-specific beta 1 glycoprotein (PSG1) is secreted from trophoblast cells of the human placenta in increasing concentrations as pregnancy progresses, becoming one of the most abundant proteins in maternal serum in the third trimester. PSG1 has seven potential N-linked glycosylation sites across its four domains. We carried out glycomic and glycoproteomic studies to characterize the glycan composition of PSG1 purified from serum of pregnant women and identified the presence of complex N-glycans containing poly LacNAc epitopes with α2,3 sialyation at four sites. Using different techniques, we explored whether PSG1 can bind to galectin-1 (Gal-1) as these two proteins were previously shown to participate in processes required for a successful pregnancy. We confirmed that PSG1 binds to Gal-1 in a carbohydrate-dependent manner with an affinity of the interaction of 0.13 µM. In addition, we determined that out of the three N-glycosylation-carrying domains, only the N and A2 domains of recombinant PSG1 interact with Gal-1. Lastly, we observed that the interaction between PSG1 and Gal-1 protects this lectin from oxidative inactivation and that PSG1 competes the ability of Gal-1 to bind to some but not all of its glycoprotein ligands.


Asunto(s)
Galectina 1/metabolismo , Polisacáridos/metabolismo , Glicoproteínas beta 1 Específicas del Embarazo/metabolismo , Femenino , Galectina 1/química , Humanos , Ligandos , Polisacáridos/química , Embarazo , Glicoproteínas beta 1 Específicas del Embarazo/química , Glicoproteínas beta 1 Específicas del Embarazo/aislamiento & purificación
8.
Methods Mol Biol ; 2132: 225-239, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32306331

RESUMEN

F-type lectins (FTLs) are characterized by a fucose recognition domain (F-type lectin domain; FTLD) that displays a novel jellyroll fold ("F-type" fold) and unique carbohydrate- and calcium-binding sequence motifs. This novel lectin family comprises widely distributed proteins exhibiting single, double, or greater multiples of the FTLD, either tandemly arrayed or combined with other structurally and functionally distinct domains. Further, differences in carbohydrate specificity among tandemly arrayed FTLDs present in any FTL polypeptide subunit, together with the expression of multiple FTL isoforms in a single individual supports a striking diversity in ligand recognition. Functions of FTLs in self/nonself recognition include innate immunity, fertilization, microbial adhesion, and pathogenesis, among others, revealing an extensive structural/functional diversification. The taxonomic distribution of FTLDs is surprisingly discontinuous, suggesting that this lectin family has been subject to secondary loss, lateral transfer, and functional co-option along evolutionary lineages.


Asunto(s)
Fucosa/metabolismo , Lectinas/química , Lectinas/metabolismo , Sitios de Unión , Secuencia de Carbohidratos , Evolución Molecular , Lectinas/genética , Modelos Moleculares , Familia de Multigenes , Unión Proteica , Conformación Proteica , Dominios Proteicos
9.
Methods Mol Biol ; 2132: 241-255, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32306332

RESUMEN

The purification of fucose-binding lectins from the liver of striped bass (Morone saxatilis), a teleost fish, and the identification of a novel lectin sequence motif led to the identification of a new family of lectins, the F-type lectins (FTLs) (see overview of the FTL family in Chapter 23 ). Isolation and purification of these proteins from liver extracts of striped bass was accomplished by affinity chromatography and size exclusion, and their identification as FTLs, by direct Edman sequencing, and protein, transcript, and gene sequence analysis. The development of specific antibodies against the M. saxatilis FTL provided an additional tool for the identification of FTLs. These methods have been successfully used for the purification of the FTL family members from tissues and body fluids of various animal species. Production and characterization of FTLs has been facilitated by the expression of the recombinant proteins. In this chapter, the biochemical characterization of FTLs is focused on the analysis of their carbohydrate specificity.


Asunto(s)
Lubina/metabolismo , Lectinas/genética , Lectinas/aislamiento & purificación , Hígado/metabolismo , Animales , Lubina/genética , Cromatografía de Afinidad , Cromatografía en Gel , Proteínas de Peces/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Lectinas/metabolismo , Familia de Multigenes , Proteínas Recombinantes/metabolismo
10.
Front Chem ; 8: 98, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32161746

RESUMEN

Both vertebrates and invertebrates display active innate immune mechanisms for defense against microbial infection, including diversified repertoires of soluble and cell-associated lectins that can effect recognition and binding to potential pathogens, and trigger downstream effector pathways that clear them from the host internal milieu. Galectins are widely distributed and highly conserved lectins that have key regulatory effects on both innate and adaptive immune responses. In addition, galectins can bind to exogenous ("non-self") carbohydrates on the surface of bacteria, enveloped viruses, parasites, and fungi, and function as recognition receptors and effector factors in innate immunity. Like most invertebrates, eastern oysters (Crassostrea virginica) and softshell clams (Mya arenaria) can effectively respond to most immune challenges through soluble and hemocyte-associated lectins. The protozoan parasite Perkinsus marinus, however, can infect eastern oysters and cause "Dermo" disease, which is highly detrimental to both natural and farmed oyster populations. The sympatric Perkinsus chesapeaki, initially isolated from infected M. arenaria clams, can also be present in oysters, and there is little evidence of pathogenicity in either clams or oysters. In this review, we discuss selected observations from our studies on the mechanisms of Perkinsus recognition that are mediated by galectin-carbohydrate interactions. We identified in the oyster two galectins that we designated CvGal1 and CvGal2, which strongly recognize P. marinus trophozoites. In the clam we also identified galectin sequences, and focused on one (that we named MaGal1) that also recognizes Perkinsus species. Here we describe the biochemical characterization of CvGal1, CvGal2, and MaGal1 with focus on the detailed study of the carbohydrate specificity, and the glycosylated moieties on the surfaces of the oyster hemocytes and the two Perkinsus species (P. marinus and P. chesapeaki). Our goal is to gain further understanding of the biochemical basis for the interactions that lead to recognition and opsonization of the Perkinsus trophozoites by the bivalve hemocytes. These basic studies on the biology of host-parasite interactions may contribute to the development of novel intervention strategies for parasitic diseases of biomedical interest.

11.
Dev Comp Immunol ; 78: 61-70, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28939483

RESUMEN

LvMKK4, a homologue of the mammalian mitogen-activated protein kinase kinase 4 (MKK4), was isolated and identified from Litopenaeus vannamei in the present study. The full-length cDNA of LvMKK4 is 1947 bp long, with an open reading frame (ORF) of 1185 bp encoding a putative protein of 388 amino acids. LvMKK4 contains several characteristic domains such as D domain, SIAKT motif and kinase domain, all of which are conserved in MAP kinase kinase family. Like mammalian MKK4 but not Drosophila MKK4, LvMKK4 could bind to, phosphorylate and activate p38 MAPK, which provided some insights into the signal transduction mechanism of MKK4-p38 cascade in invertebrates. Our real-time PCR data indicated that LvMKK4 was ubiquitously expressed in all tested tissues and extraordinarily abundant in muscle. Dual luciferase reporter assays in Drosophila S2 cells revealed that LvMKK4 activated the transcription of antimicrobial peptide genes (AMPs), including Drosophila Attacin A, Drosomycin, and shrimp Penaeidins. Additionally, LvMKK4 was up-regulated in both intestine and hepatopancreas by a variety of inflammatory stimuli including LPS, Vibrio parahaemolyticus, Staphhylococcu saureus, Poly (I: C) and white spot syndrome virus. Furthermore, RNAi-mediated knockdown of LvMKK4 enhanced the sensitivity of L. vannamei to V. parahaemolyticus infection. These findings suggested that LvMKK4 played an important role in anti-bacterial response and could be a potential target for inflammation treatment.


Asunto(s)
Antibacterianos/metabolismo , Proteínas de Artrópodos/metabolismo , Infecciones por Virus ADN/inmunología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Penaeidae/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Vibriosis/inmunología , Vibrio parahaemolyticus/inmunología , Virus del Síndrome de la Mancha Blanca 1/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Proteínas de Artrópodos/genética , Línea Celular , Clonación Molecular , Drosophila/genética , Drosophila/inmunología , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/genética , MAP Quinasa Quinasa 4/genética , Mamíferos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Penaeidae/genética , Poli I-C/inmunología , Unión Proteica , ARN Interferente Pequeño/genética
12.
Front Immunol ; 8: 1648, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29238345

RESUMEN

The F-type lectin (FTL) family is one of the most recent to be identified and structurally characterized. Members of the FTL family are characterized by a fucose recognition domain [F-type lectin domain (FTLD)] that displays a novel jellyroll fold ("F-type" fold) and unique carbohydrate- and calcium-binding sequence motifs. This novel lectin family comprises widely distributed proteins exhibiting single, double, or greater multiples of the FTLD, either tandemly arrayed or combined with other structurally and functionally distinct domains, yielding lectin subunits of pleiotropic properties even within a single species. Furthermore, the extraordinary variability of FTL sequences (isoforms) that are expressed in a single individual has revealed genetic mechanisms of diversification in ligand recognition that are unique to FTLs. Functions of FTLs in self/non-self-recognition include innate immunity, fertilization, microbial adhesion, and pathogenesis, among others. In addition, although the F-type fold is distinctive for FTLs, a structure-based search revealed apparently unrelated proteins with minor sequence similarity to FTLs that displayed the FTLD fold. In general, the phylogenetic analysis of FTLD sequences from viruses to mammals reveals clades that are consistent with the currently accepted taxonomy of extant species. However, the surprisingly discontinuous distribution of FTLDs within each taxonomic category suggests not only an extensive structural/functional diversification of the FTLs along evolutionary lineages but also that this intriguing lectin family has been subject to frequent gene duplication, secondary loss, lateral transfer, and functional co-option.

13.
mBio ; 8(3)2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28655817

RESUMEN

Neuraminidases (NAs) are critical virulence factors for several microbial pathogens. With a highly conserved catalytic domain, a microbial NA "superfamily" has been proposed. We previously reported that murine polymorphonuclear leukocyte (PMN) sialidase activity was important in leukocyte trafficking to inflamed sites and that antibodies to Clostridium perfringens NA recognized a cell surface molecule(s), presumed to be a sialidase of eukaryotic origin on interleukin-8-stimulated human and murine PMNs. These antibodies also inhibited cell sialidase activity both in vitro and, in the latter instance, in vivo We therefore hypothesized that mammalian sialidases share structural homology and epitopes with microbial NAs. We now report that antibodies to one of the isoforms of C. perfringens NA, as well as anti-influenza virus NA serum, recognize human NEU3 but not NEU1 and that antibodies to C. perfringens NA inhibit NEU3 enzymatic activity. We conclude that the previously described microbial NA superfamily extends to human sialidases. Strategies designed to therapeutically inhibit microbial NA may need to consider potential compromising effects on human sialidases, particularly those expressed in cells of the immune system.IMPORTANCE We previously reported that sialidase activity of human neutrophils plays a critical role in the host inflammatory response. Since the catalytic domains of microbial neuraminidases are highly conserved, we hypothesized that antibodies against Clostridium perfringens neuraminidase might inhibit mammalian sialidase activity. Before the recognition of four mammalian sialidase (Neu) isoforms, we demonstrated that anti-C. perfringens neuraminidase antibodies inhibited human and murine sialidase activity in vivo and in vitro We now show that the antibodies to microbial neuraminidase (C. perfringens and influenza virus) recognize human NEU3, which is important for neural development and cell signaling. Since many microbes that infect mucosal surfaces express neuraminidase, it is possible that the use of sialidase inhibitors (e.g., zanamivir), might also compromise human sialidase activity critical to the human immune response. Alternatively, sialidase inhibitors may prove useful in the treatment of hyperinflammatory conditions.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Antivirales/inmunología , Clostridium perfringens/inmunología , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/inmunología , Proteínas Virales/inmunología , Humanos
14.
Pathog Dis ; 75(5)2017 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-28449072

RESUMEN

Carbohydrate structures on the cell surface encode complex information that through specific recognition by carbohydrate-binding proteins (lectins) modulates interactions between cells, cells and the extracellular matrix, or mediates recognition of potential microbial pathogens. Galectins are a family of ß-galactoside-binding lectins, which are evolutionary conserved and have been identified in most organisms, from fungi to invertebrates and vertebrates, including mammals. Since their discovery in the 1970s, their biological roles, initially understood as limited to recognition of endogenous carbohydrate ligands in embryogenesis and development, have expanded in recent years by the discovery of their roles in tissue repair and regulation of immune homeostasis. More recently, evidence has accumulated to support the notion that galectins can also bind glycans on the surface of potentially pathogenic microbes, and function as recognition and effector factors in innate immunity, thus establishing a new paradigm. Furthermore, some parasites 'subvert' the recognition roles of the vector/host galectins for successful attachment or invasion. These recent findings have revealed a striking functional diversification in this structurally conserved lectin family.


Asunto(s)
Galectinas/metabolismo , Interacciones Huésped-Patógeno , Evasión Inmune , Inmunidad Innata , Receptores Inmunológicos/metabolismo , Animales , Humanos
15.
Dev Comp Immunol ; 55: 241-252, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26429411

RESUMEN

The infectious hematopoietic necrosis virus (IHNV; Rhabdoviridae, Novirhabdovirus) infects teleost fish, such as salmon and trout, and is responsible for significant losses in the aquaculture industry and in wild fish populations. Although IHNV enters the host through the skin at the base of the fins, the viral adhesion and entry mechanisms are not fully understood. In recent years, evidence has accumulated in support of the key roles played by protein-carbohydrate interactions between host lectins secreted to the extracellular space and virion envelope glycoproteins in modulating viral adhesion and infectivity. In this study, we assessed in vitro the potential role(s) of zebrafish (Danio rerio) proto type galectin-1 (Drgal1-L2) and a chimera galectin-3 (Drgal3-L1) in IHNV adhesion to epithelial cells. Our results suggest that the extracellular Drgal1-L2 and Drgal3-L1 interact directly and in a carbohydrate-dependent manner with the IHNV glycosylated envelope and glycans on the epithelial cell surface, significantly reducing viral adhesion.


Asunto(s)
Células Epiteliales/fisiología , Galectinas/metabolismo , Virus de la Necrosis Hematopoyética Infecciosa/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Infecciones por Rhabdoviridae/inmunología , Proteínas del Envoltorio Viral/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/inmunología , Animales , Células Cultivadas , Células Epiteliales/virología , Galectinas/genética , Interacciones Huésped-Patógeno , Virus de la Necrosis Hematopoyética Infecciosa/patogenicidad , Proteínas Recombinantes de Fusión/genética , Infecciones por Rhabdoviridae/transmisión , Virulencia , Acoplamiento Viral , Proteínas de Pez Cebra/genética
16.
Mol Immunol ; 68(2 Pt A): 194-202, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26355912

RESUMEN

Influenza patients frequently display increased susceptibility to Streptococcus pneumoniae co-infection and sepsis, the prevalent cause of mortality during influenza pandemics. However, the detailed mechanisms by which an influenza infection predisposes patients to suffer pneumococcal pneumonia are not fully understood. A murine model for influenza infection closely reflects the observations in human patients, since if the animals that have recovered from influenza A virus (IAV) sublethal infection are challenged with S. pneumoniae, they undergo a usually fatal uncontrolled cytokine response. We have previously demonstrated both in vitro and in vivo that the expression and secretion of galectin-1 (Gal1) and galectin-3 (Gal3) are modulated during IAV infection, and that the viral neuraminidase unmasks galactosyl moieties in the airway epithelia. In this study we demonstrate in vitro that the binding of secreted Gal1 and Gal3 to the epithelial cell surface modulates the expression of SOCS1 and RIG1, and activation of ERK, AKT or JAK/STAT1 signaling pathways, leading to a disregulated expression and release of pro-inflammatory cytokines. Our results suggest that the activity of the viral and pneumococcal neuraminidases on the surface of the airway epithelial cells function as a "danger signal" that leads to rapid upregulation of SOCS1 expression to prevent an uncontrolled inflammatory response. The binding of extracellular Gal1 or Gal3 to the galactosyl moieties unmasked on the surface of airway epithelial cells can either "fine-tune" or severely disregulate this process, respectively, the latter potentially leading to hypercytokinemia.


Asunto(s)
ARN Helicasas DEAD-box/genética , Células Epiteliales/inmunología , Galectina 1/farmacología , Galectina 3/farmacología , Proteínas Supresoras de la Señalización de Citocinas/genética , Animales , Proteínas Bacterianas/farmacología , Línea Celular Tumoral , Citocinas/biosíntesis , Citocinas/metabolismo , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Galectina 1/biosíntesis , Galectina 1/inmunología , Galectina 3/biosíntesis , Galectina 3/inmunología , Regulación de la Expresión Génica , Humanos , Inflamación , Virus de la Influenza A/inmunología , Quinasas Janus/genética , Quinasas Janus/inmunología , Ratones , Neuraminidasa/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptores Inmunológicos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Proteínas Supresoras de la Señalización de Citocinas/farmacología
17.
Biochemistry ; 54(30): 4711-30, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26158802

RESUMEN

Galectins are highly conserved lectins that are key to multiple biological functions, including pathogen recognition and regulation of immune responses. We previously reported that CvGal1, a galectin expressed in phagocytic cells (hemocytes) of the eastern oyster (Crassostrea virginica), is hijacked by the parasite Perkinsus marinus to enter the host, where it causes systemic infection and death. Screening of an oyster hemocyte cDNA library revealed a novel galectin, which we designated CvGal2, with four tandemly arrayed carbohydrate recognition domains (CRDs). Phylogentic analysis of the CvGal2 CRDs suggests close relationships with homologous CRDs from CvGal1. Glycan array analysis, however, revealed that, unlike CvGal1 which preferentially binds to the blood group A tetrasaccharide, CvGal2 recognizes both blood group A and B tetrasaccharides and related structures, suggesting that CvGal2 has broader binding specificity. Furthermore, SPR analysis demonstrated significant differences in the binding kinetics of CvGal1 and CvGal2, and structural modeling revealed substantial differences in their interactions with the oligosaccharide ligands. CvGal2 is homogeneously distributed in the hemocyte cytoplasm, is released to the extracellular space, and binds to the hemocyte surface. CvGal2 binds to P. marinus trophozoites in a dose-dependent and ß-galactoside-specific manner. Strikingly, negligible binding of CvGal2 was observed for Perkinsus chesapeaki, a sympatric parasite species mostly prevalent in the clams Mya arenaria and Macoma balthica. The differential recognition of Perkinsus species by the oyster galectins is consistent with their relative prevalence in oyster and clam species and supports their role in facilitating parasite entry and infectivity in a host-preferential manner.


Asunto(s)
Alveolados , Antígenos de Grupos Sanguíneos , Crassostrea , Galectinas , Oligosacáridos , Filogenia , Alveolados/química , Alveolados/genética , Alveolados/metabolismo , Animales , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Crassostrea/química , Crassostrea/genética , Crassostrea/metabolismo , Crassostrea/parasitología , Galectinas/química , Galectinas/genética , Galectinas/metabolismo , Hemocitos/química , Hemocitos/metabolismo , Hemocitos/parasitología , Oligosacáridos/química , Oligosacáridos/genética , Oligosacáridos/metabolismo
18.
Mol Immunol ; 65(1): 1-16, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25597246

RESUMEN

The continued threat of worldwide influenza pandemics, together with the yearly emergence of antigenically drifted influenza A virus (IAV) strains, underscore the urgent need to elucidate not only the mechanisms of influenza virulence, but also those mechanisms that predispose influenza patients to increased susceptibility to subsequent infection with Streptococcus pneumoniae. Glycans displayed on the surface of epithelia that are exposed to the external environment play important roles in microbial recognition, adhesion, and invasion. It is well established that the IAV hemagglutinin and pneumococcal adhesins enable their attachment to the host epithelia. Reciprocally, the recognition of microbial glycans by host carbohydrate-binding proteins (lectins) can initiate innate immune responses, but their relevance in influenza or pneumococcal infections is poorly understood. Galectins are evolutionarily conserved lectins characterized by affinity for ß-galactosides and a unique sequence motif, with critical regulatory roles in development and immune homeostasis. In this study, we examined the possibility that galectins expressed in the airway epithelial cells might play a significant role in viral or pneumococcal adhesion to airway epithelial cells. Our results in a mouse model for influenza and pneumococcal infection revealed that the murine lung expresses a diverse galectin repertoire, from which selected galectins, including galectin 1 (Gal1) and galectin 3 (Gal3), are released to the bronchoalveolar space. Further, the results showed that influenza and subsequent S. pneumoniae infections significantly alter the glycosylation patterns of the airway epithelial surface and modulate galectin expression. In vitro studies on the human airway epithelial cell line A549 were consistent with the observations made in the mouse model, and further revealed that both Gal1 and Gal3 bind strongly to IAV and S. pneumoniae, and that exposure of the cells to viral neuraminidase or influenza infection increased galectin-mediated S. pneumoniae adhesion to the cell surface. Our results suggest that upon influenza infection, pneumococcal adhesion to the airway epithelial surface is enhanced by an interplay among the host galectins and viral and pneumococcal neuraminidases. The observed enhancement of pneumococcal adhesion may be a contributing factor to the observed hypersusceptibility to pneumonia of influenza patients.


Asunto(s)
Galectina 1/metabolismo , Galectina 3/metabolismo , Infecciones por Orthomyxoviridae/patología , Infecciones Neumocócicas/patología , Mucosa Respiratoria/citología , Adhesinas Bacterianas , Animales , Apoptosis , Adhesión Bacteriana/fisiología , Línea Celular , Susceptibilidad a Enfermedades , Células Epiteliales/metabolismo , Galectina 1/biosíntesis , Galectina 3/biosíntesis , Glicoproteínas Hemaglutininas del Virus de la Influenza , Humanos , Virus de la Influenza A/patogenicidad , Ratones , Ratones Endogámicos C57BL , Neuraminidasa/farmacología , Unión Proteica/efectos de los fármacos , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/virología , Streptococcus pneumoniae/patogenicidad
19.
Methods Mol Biol ; 1207: 327-41, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25253151

RESUMEN

Techniques for disrupting gene expression are invaluable tools for the analysis of the biological role(s) of a gene product. Because of its genetic tractability and multiple advantages over conventional mammalian models, the zebrafish (Danio rerio) is recognized as a powerful system for gaining new insight into diverse aspects of human health and disease. Among the multiple mammalian gene families for which the zebrafish has shown promise as an invaluable model for functional studies, the galectins have attracted great interest due to their participation in early development, regulation of immune homeostasis, and recognition of microbial pathogens. Galectins are ß-galactosyl-binding lectins with a characteristic sequence motif in their carbohydrate recognition domains (CRDs), which comprise an evolutionary conserved family ubiquitous in eukaryotic taxa. Galectins are emerging as key players in the modulation of many important pathological processes, which include acute and chronic inflammatory diseases, autoimmunity and cancer, thus making them potential molecular targets for innovative drug discovery. Here, we provide a review of the current methods available for the manipulation of gene expression in the zebrafish, with a focus on gene knockdown [morpholino (MO)-derived antisense oligonucleotides] and knockout (CRISPR-Cas) technologies.


Asunto(s)
Galectinas/genética , Regulación de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen/métodos , Técnicas de Inactivación de Genes/métodos , Pez Cebra/genética , Animales , Secuencia de Bases , Embrión no Mamífero , Femenino , Galectinas/deficiencia , Inyecciones , Masculino , Morfolinos/genética , Fenotipo , ARN/genética , Pez Cebra/embriología
20.
Am J Physiol Lung Cell Mol Physiol ; 306(9): L876-86, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24658138

RESUMEN

Sialic acids on glycoconjugates play a pivotal role in many biological processes. In the airways, sialylated glycoproteins and glycolipids are strategically positioned on the plasma membranes of epithelia to regulate receptor-ligand, cell-cell, and host-pathogen interactions at the molecular level. We now demonstrate, for the first time, sialidase activity for ganglioside substrates in human airway epithelia. Of the four known mammalian sialidases, NEU3 has a substrate preference for gangliosides and is expressed at mRNA and protein levels at comparable abundance in epithelia derived from human trachea, bronchi, small airways, and alveoli. In small airway and alveolar epithelia, NEU3 protein was immunolocalized to the plasma membrane, cytosolic, and nuclear subcellular fractions. Small interfering RNA-induced silencing of NEU3 expression diminished sialidase activity for a ganglioside substrate by >70%. NEU3 immunostaining of intact human lung tissue could be localized to the superficial epithelia, including the ciliated brush border, as well as to nuclei. However, NEU3 was reduced in subepithelial tissues. These results indicate that human airway epithelia express catalytically active NEU3 sialidase.


Asunto(s)
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Epitelio/metabolismo , Neuraminidasa/metabolismo , Sistema Respiratorio/metabolismo , Biotinilación , Western Blotting , Catálisis , Células Cultivadas , Citometría de Flujo , Gangliósidos/metabolismo , Humanos , Técnicas para Inmunoenzimas , Neuraminidasa/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ácidos Siálicos/metabolismo , Fracciones Subcelulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...