Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 298(12): 102591, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36244450

RESUMEN

The macrophage mannose receptor (MR, CD206) is a transmembrane endocytic lectin receptor, expressed in selected immune and endothelial cells, and is involved in immunity and maintaining homeostasis. Eight of the ten extracellular domains of the MR are C-type lectin domains (CTLDs) which mediate the binding of mannose, fucose, and GlcNAc in a calcium-dependent manner. Previous studies indicated that self-glycosylation of MR regulates its glycan binding. To further explore this structure-function relationship, we studied herein a recombinant version of mouse MR CTLD4-7 fused to human Fc-portion of IgG (MR-Fc). The construct was expressed in different glycosylation-mutant cell lines to study the influence of differential glycosylation on receptor glycan-binding properties. We conducted site-specific N- and O-glycosylation analysis and glycosylation site characterization using mass spectrometry by which several novel O-glycosylation sites were identified in mouse MR and confirmed in human full-length MR. This information guided experiments evaluating the receptor functionality by glycan microarray analysis in combination with glycan-modifying enzymes. Treatment of active MR-Fc with combinations of exoglycosidases, including neuraminidase and galactosidases, resulted in the loss of trans-binding (binding of MR CTLDs to non-MR glycans), due to unmasking of terminal, nonreducing GlcNAc in N-glycans of the MR CTLDs. Regalactosylation of N-glycans rescues mannose binding by MR-Fc. Our results indicate that glycans within the MR CTLDs act as a regulatory switch by masking and unmasking self-ligands, including terminal, nonreducing GlcNAc in N-glycans, which could control MR activity in a tissue- and cell-specific manner or which potentially affect bacterial pathogenesis in an immunomodulatory fashion.


Asunto(s)
Lectinas Tipo C , Receptor de Manosa , Humanos , Animales , Ratones , Lectinas Tipo C/metabolismo , Glicosilación , Manosa , Células Endoteliales/metabolismo , Polisacáridos/metabolismo
2.
Cell Mol Life Sci ; 78(14): 5569-5585, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34089345

RESUMEN

The macrophage mannose receptor (CD206, MR) is an endocytic lectin receptor which plays an important role in homeostasis and innate immunity, however, the endogenous glycan and glycoprotein ligands recognized by its C-type lectin domains (CTLD) have not been well studied. Here we used the murine MR CTLD4-7 coupled to the Fc-portion of human IgG (MR-Fc) to investigate the MR glycan and glycoprotein recognition. We probed 16 different cancer and control tissues using the MR-Fc, and observed cell- and tissue-specific binding with varying intensity. All cancer tissues and several control tissues exhibited MR-Fc ligands, intracellular and/or surface-located. We further confirmed the presence of ligands on the surface of cancer cells by flow cytometry. To characterize the fine specificity of the MR for glycans, we screened a panel of glycan microarrays. Remarkably, the results indicate that the CTLD4-7 of the MR is highly selective for specific types of pauci- and oligomannose N-glycans among hundreds of glycans tested. As lung cancer tissue and the lung cancer cell line A549 showed intense MR-Fc binding, we further investigated the MR glycoprotein ligands in those cells by immunoprecipitation and glycoproteomic analysis. All enriched glycoproteins, of which 42 were identified, contained pauci- or oligomannose N-glycans, confirming the microarray results. Our study demonstrates that the MR CTLD4-7 is highly selective for pauci- and oligomannosidic N-glycans, structures that are often elevated in tumor cells, and suggest a potential role for the MR in tumor biology.


Asunto(s)
Glicoproteínas/metabolismo , Lectinas Tipo C/metabolismo , Neoplasias Pulmonares/patología , Lectinas de Unión a Manosa/metabolismo , Oligosacáridos/metabolismo , Polisacáridos/metabolismo , Receptores de Superficie Celular/metabolismo , Células A549 , Glicoproteínas/genética , Glicosilación , Humanos , Lectinas Tipo C/genética , Ligandos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Modelos Moleculares , Receptores de Superficie Celular/genética
3.
J Biol Chem ; 293(27): 10646-10662, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29769321

RESUMEN

Innate immunity is critical in the early containment of influenza A virus (IAV) infection, and surfactant protein D (SP-D) plays a crucial role in the pulmonary defense against IAV. In pigs, which are important intermediate hosts during the generation of pandemic IAVs, SP-D uses its unique carbohydrate recognition domain (CRD) to interact with IAV. An N-linked CRD glycosylation provides interactions with the sialic acid-binding site of IAV, and a tripeptide loop at the lectin-binding site facilitates enhanced interactions with IAV glycans. Here, to investigate both mechanisms of IAV neutralization in greater detail, we produced an N-glycosylated neck-CRD fragment of porcine SP-D (RpNCRD) in HEK293 cells. X-ray crystallography disclosed that the N-glycan did not alter the CRD backbone structure, including the lectin site conformation, but revealed a potential second nonlectin-binding site for glycans. IAV hemagglutination inhibition, IAV aggregation, and neutralization of IAV infection studies showed that RpNCRD, unlike the human analogue RhNCRD, exhibits potent neutralizing activity against pandemic A/Aichi/68 (H3N2), enabled by both porcine-specific structural features of its CRD. MS analysis revealed an N-glycan site-occupancy of >98% at Asn-303 of RpNCRD with complex-type, heterogeneously branched and predominantly α(2,3)-sialylated oligosaccharides. Glycan-binding array data characterized both RpNCRD and RhNCRD as mannose-type lectins. RpNCRD also bound LewisY structures, whereas RhNCRD bound polylactosamine-containing glycans. The presence of the N-glycan in the CRD increases the glycan-binding specificity of RpNCRD. These insights increase our understanding of porcine-specific innate defense against pandemic IAV and may inform the design of recombinant SP-D-based antiviral drugs.


Asunto(s)
Inmunidad Innata/inmunología , Virus de la Influenza A/inmunología , Lectinas/metabolismo , Infecciones por Orthomyxoviridae/prevención & control , Polisacáridos/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Ácidos Siálicos/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Conformación de Carbohidratos , Glicosilación , Pruebas de Inhibición de Hemaglutinación , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Polisacáridos/química , Proteína D Asociada a Surfactante Pulmonar/química , Proteína D Asociada a Surfactante Pulmonar/genética , Homología de Secuencia , Porcinos
4.
J Infect Dis ; 217(2): 310-319, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29136163

RESUMEN

Ascaris suum is a helminth parasite of pigs closely related to its human counterpart, A. lumbricoides, which infects almost 1 billion people. Ascaris is thought to modulate host immune and inflammatory responses, which may drive immune hyporesponsiveness during chronic infections. Using transcriptomic analysis, we show here that pigs with a chronic A. suum infection have a substantial suppression of inflammatory pathways in the intestinal mucosa, with a broad downregulation of genes encoding cytokines and antigen-processing and costimulatory molecules. A. suum body fluid (ABF) suppressed similar transcriptional pathways in human dendritic cells (DCs) in vitro. DCs exposed to ABF secreted minimal amounts of cytokines and had impaired production of cyclooxygengase-2, altered glucose metabolism, and reduced capacity to induce interferon-gamma production in T cells. Our in vivo and in vitro data provide an insight into mucosal immune modulation during Ascaris infection, and show that A. suum profoundly suppresses immune and inflammatory pathways.


Asunto(s)
Ascariasis/patología , Ascaris suum/inmunología , Células Dendríticas/inmunología , Tolerancia Inmunológica , Mucosa Intestinal/patología , Animales , Ascariasis/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Mucosa Intestinal/inmunología , Modelos Biológicos , Porcinos
5.
FASEB J ; 31(2): 719-731, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27806992

RESUMEN

Clinical trials have shown that administration of the nematode Trichuris suis can be beneficial in treating various immune disorders. To provide insight into the mechanisms by which this worm suppresses inflammatory responses, an active component was purified from T. suis soluble products (TsSPs) that suppress---- TNF and IL-12 secretion from LPS-activated human dendritic cells (DCs). Analysis by liquid chromatography tandem mass spectrometry identified this compound as prostaglandin (PG)E2. The purified compound showed similar properties compared with TsSPs and commercial PGE2 in modulating LPS-induced expression of many cytokines and chemokines and in modulating Rab7B and P2RX7 expression in human DCs. Furthermore, the TsSP-induced reduction of TNF secretion from DCs is reversed by receptor antagonists for EP2 and EP4, indicating PGE2 action. T. suis secretes extremely high amounts of PGE2 (45-90 ng/mg protein) within their excretory/secretory products but few related lipid mediators as established by metabololipidomic analysis. Culture of T. suis with several cyclooxygenase (COX) inhibitors that inhibit mammalian prostaglandin synthesis affected the worm's motility but did not inhibit PGE2 secretion, suggesting that the worms can synthesize PGE2 via a COX-independent pathway. We conclude that T. suis secretes PGE2 to suppress proinflammatory responses in human DCs, thereby modulating the host's immune response.-Laan, L. C., Williams, A. R., Stavenhagen, K., Giera, M., Kooij, G., Vlasakov, I., Kalay, H., Kringel, H., Nejsum, P., Thamsborg, S. M., Wuhrer, M., Dijkstra, C. D., Cummings, R. D., van Die, I. The whipworm (Trichuris suis) secretes prostaglandin E2 to suppress proinflammatory properties in human dendritic cells.


Asunto(s)
Células Dendríticas/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacología , Inflamación/metabolismo , Trichuris/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lipopolisacáridos/toxicidad , Estructura Molecular , Especificidad de la Especie
6.
Immunology ; 150(3): 312-328, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27905107

RESUMEN

Interactions between dendritic cells (DCs) and environmental, dietary and pathogen antigens play a key role in immune homeostasis and regulation of inflammation. Dietary polyphenols such as proanthocyanidins (PAC) may reduce inflammation, and we therefore hypothesized that PAC may suppress lipopolysaccharide (LPS) -induced responses in human DCs and subsequent T helper type 1 (Th1) -type responses in naive T cells. Moreover, we proposed that, because DCs are likely to be exposed to multiple stimuli, the activity of PAC may synergise with other bioactive molecules that have anti-inflammatory activity, e.g. soluble products from the helminth parasite Trichuris suis (TsSP). We show that PAC are endocytosed by monocyte-derived DCs and selectively induce CD86 expression. Subsequently, PAC suppress the LPS-induced secretion of interleukin-6 (IL-6) and IL-12p70, while enhancing secretion of IL-10. Incubation of DCs with PAC did not affect lymphocyte proliferation; however, subsequent interferon-γ production was markedly suppressed, while IL-4 production was unaffected. The activity of PAC was confined to oligomers (degree of polymerization ≥ 4). Co-pulsing DCs with TsSP and PAC synergistically reduced secretion of tumour necrosis factor-α, IL-6 and IL-12p70 while increasing IL-10 secretion. Moreover, both TsSP and PAC alone induced Th2-associated OX40L expression in DCs, and together synergized to up-regulate OX40L. These data suggest that PAC induce an anti-inflammatory phenotype in human DCs that selectively down-regulates Th1 response in naive T cells, and that they also act cooperatively with TsSP. Our results indicate a novel interaction between dietary compounds and parasite products to influence immune function, and may suggest that combinations of PAC and TsSP can have therapeutic potential for inflammatory disorders.


Asunto(s)
Antiinflamatorios/farmacología , Células Dendríticas/efectos de los fármacos , Inflamación/tratamiento farmacológico , Proantocianidinas/farmacología , Células TH1/inmunología , Células Th2/inmunología , Tricuriasis/tratamiento farmacológico , Animales , Antígenos Helmínticos/inmunología , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/parasitología , Quimioterapia Combinada , Humanos , Inflamación/inmunología , Activación de Linfocitos , Ligando OX40/genética , Ligando OX40/metabolismo , Interferencia de ARN , Porcinos , Balance Th1 - Th2/efectos de los fármacos , Tricuriasis/inmunología , Trichuris/inmunología , Regulación hacia Arriba
7.
FASEB J ; 30(8): 2826-36, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27095802

RESUMEN

Helminths have strong immunoregulatory properties that may be exploited in treatment of chronic immune disorders, such as multiple sclerosis and inflammatory bowel disease. Essential players in the pathogenesis of these diseases are proinflammatory macrophages. We present evidence that helminths modulate the function and phenotype of these innate immune cells. We found that soluble products derived from the Trichuris suis (TsSP) significantly affect the differentiation of monocytes into macrophages and their subsequent polarization. TsSPs reduce the expression and production of inflammatory cytokines, including IL-6 and TNF, in human proinflammatory M1 macrophages. TsSPs induce a concomitant anti-inflammatory M2 signature, with increased IL-10 production. Furthermore, they suppress CHIT activity and enhance secretion of matrix metalloproteinase 9. Short-term triggering of monocytes with TsSPs early during monocyte-to-macrophage differentiation imprinted these phenotypic alterations, suggesting long-lasting epigenetic changes. The TsSP-induced effects in M1 macrophages were completely reversed by inhibiting histone deacetylases, which corresponded with decreased histone acetylation at the TNF and IL6 promoters. These results demonstrate that TsSPs have a potent and sustained immunomodulatory effect on human macrophage differentiation and polarization through epigenetic remodeling and provide new insights into the mechanisms by which helminths modulate human immune responses.-Hoeksema, M. A., Laan, L. C., Postma, J. J., Cummings, R. D., de Winther, M. P. J., Dijkstra, C. D., van Die, I., Kooij, G. Treatment with Trichuris suis soluble products during monocyte-to-macrophage differentiation reduces inflammatory responses through epigenetic remodeling.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Lipopolisacáridos/farmacología , Macrófagos/fisiología , Monocitos/fisiología , Trichuris/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Epigénesis Genética/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Helminto , Humanos , Inflamación , Lipopolisacáridos/química , Trichuris/química
8.
J Biol Chem ; 290(31): 19018-33, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26063800

RESUMEN

The T-synthase (core 1 ß3-galactosyltransferase) and its molecular chaperone Cosmc regulate the biosynthesis of mucin type O-glycans on glycoproteins, and evidence suggests that both T-synthase and Cosmc are transcriptionally suppressed in several human diseases, although the transcriptional regulation of these two genes is not understood. Here, we characterized the promoters essential for human Cosmc and T-synthase transcription. The upstream regions of the genes lack a conventional TATA box but contain CpG islands, cCpG-I and cCpG-II for Cosmc and tCpG for T-synthase. Using luciferase reporter assays, site-directed mutagenesis, ChIP assays, and mithramycin A treatment, we identified the core promoters within cCpG-II and tCpG, which contain two binding sites for Krüppel-like transcription factors, including SP1/SP3, respectively. Methylome analysis of Tn4 B cells, which harbor a silenced Cosmc, confirmed the hypermethylation of the Cosmc core promoter but not for T-synthase. These results demonstrate that Cosmc and T-synthase are transcriptionally regulated at a basal level by the specificity protein/Krüppel-like transcription factor family of members, which explains their ubiquitous and coordinated expression, and also indicate that they are differentially epigenetically regulated beyond X chromosome imprinting. These results are important in understanding the regulation of these genes that have roles in human diseases, such as IgA nephropathy and cancer.


Asunto(s)
Galactosiltransferasas/genética , Chaperonas Moleculares/genética , Secuencia de Bases , Sitios de Unión , Islas de CpG , Metilación de ADN , Epigénesis Genética , Secuencia Rica en GC , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/metabolismo
9.
Infect Immun ; 84(5): 1371-1386, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26883596

RESUMEN

Infection of mammals by the parasitic helminth Schistosoma mansoni induces antibodies to glycan antigens in worms and eggs, but the differential nature of the immune response among infected mammals is poorly understood. To better define these responses, we used a shotgun glycomics approach in which N-glycans from schistosome egg glycoproteins were prepared, derivatized, separated, and used to generate an egg shotgun glycan microarray. This array was interrogated with sera from infected mice, rhesus monkeys, and humans and with glycan-binding proteins and antibodies to gather information about the structures of antigenic glycans, which also were analyzed by mass spectrometry. A major glycan antigen targeted by IgG from different infected species is the FLDNF epitope [Fucα3GalNAcß4(Fucα3)GlcNAc-R], which is also recognized by the IgG monoclonal antibody F2D2. The FLDNF antigen is expressed by all life stages of the parasite in mammalian hosts, and F2D2 can kill schistosomula in vitro in a complement-dependent manner. Different antisera also recognized other glycan determinants, including core ß-xylose and highly fucosylated glycans. Thus, the natural shotgun glycan microarray of schistosome eggs is useful in identifying antigenic glycans and in developing new anti-glycan reagents that may have diagnostic applications and contribute to developing new vaccines against schistosomiasis.


Asunto(s)
Antígenos Helmínticos/inmunología , Polisacáridos/inmunología , Schistosoma mansoni/inmunología , Cigoto/inmunología , Animales , Anticuerpos Antihelmínticos/sangre , Antígenos Helmínticos/química , Antígenos Helmínticos/aislamiento & purificación , Epítopos/inmunología , Femenino , Humanos , Inmunoglobulina G/sangre , Macaca mulatta , Espectrometría de Masas , Ratones , Análisis por Micromatrices , Polisacáridos/química , Polisacáridos/aislamiento & purificación , Unión Proteica
10.
Mol Cell Proteomics ; 13(11): 3097-113, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25071157

RESUMEN

Immunoglobulin A nephropathy (IgAN) is the most common form of glomerulonephritis worldwide and is histologically characterized by the deposition of IgA1 and consequent inflammation in the glomerular mesangium. Prior studies suggested that serum IgA1 from IgAN patients contains aberrant, undergalactosylated O-glycans, for example, Tn antigen and its sialylated version, SialylTn (STn), but the mechanisms underlying aberrant O-glycosylation are not well understood. Here we have used serial lectin separation technologies, Western blot, enzymatic modifications, and mass spectrometry to explore whether there are different glycoforms of IgA1 in plasma from patients with IgAN and healthy individuals. Although total plasma IgA in IgAN patients was elevated ∼ 1.6-fold compared with that in healthy donors, IgA1 in all samples was unexpectedly separable into two distinct glycoforms: one with core 1 based O-glycans, and the other exclusively containing Tn/STn structures. Importantly, Tn antigen present on IgA1 from IgAN patients and controls was convertible into the core 1 structure in vitro by recombinant T-synthase. Our results demonstrate that undergalactosylation of O-glycans in IgA1 is not restricted to IgAN and suggest that in vivo inefficiency of T-synthase toward IgA1 in a subpopulation of B or plasma cells, as well as overall elevation of IgA, may contribute to IgAN pathogenesis.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Galactosiltransferasas/metabolismo , Glomerulonefritis por IGA/sangre , Inmunoglobulina A/sangre , Polisacáridos/metabolismo , Adulto , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Linfocitos B/inmunología , Femenino , Galactosa/metabolismo , Mesangio Glomerular/inmunología , Mesangio Glomerular/patología , Glicosilación , Humanos , Inmunoglobulina A/clasificación , Inmunoglobulina A/inmunología , Inflamación/inmunología , Lectinas/inmunología , Masculino , Aglutinina de Mani/inmunología , Polisacáridos/sangre , Sialiltransferasas/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
11.
BMC Microbiol ; 15: 253, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26526354

RESUMEN

BACKGROUND: Galactofuranose (Galf)-containing glycoconjugates are present in numerous microbes, including filamentous fungi where they are important for morphology, virulence and maintaining cell wall integrity. The incorporation of Galf-residues into galactomannan, galactomannoproteins and glycolipids is carried out by Golgi-localized Galf transferases. The nucleotide sugar donor used by these transferases (UDP-Galf) is produced in the cytoplasm and has to be transported to the lumen of the Golgi by a dedicated nucleotide sugar transporter. METHODS: Based on homology with recently identified UDP-Galf-transporters in A. fumigatus and A. nidulans, two putative UDP-Galf-transporters in A. niger were found. Their function and localization was determined by gene deletions and GFP-tagging studies, respectively. RESULTS: The two putative UDP-Galf-transporters in A. niger are homologous to each other and are predicted to contain eleven transmembrane domains (UgtA) or ten transmembrane domains (UgtB) due to a reduced length of the C-terminal part of the UgtB protein. The presence of two putative UDP-Galf-transporters in the genome was not unique for A. niger. From the twenty Aspergillus species analysed, nine species contained two additional putative UDP-Galf-transporters. Three of the nine species were outside the Aspergillus section nigri, indication an early duplication of UDP-Galf-transporters and subsequent loss of the UgtB copy in several aspergilli. Deletion analysis of the single and double mutants in A. niger indicated that the two putative UDP-Galf-transporters (named UgtA and UgtB) have a redundant function in UDP-Galf-transport as only the double mutant displayed a Galf-negative phenotype. The Galf-negative phenotype of the double mutant could be complemented by expressing either CFP-UgtA or CFP-UgtB fusion proteins from their endogenous promoters, indicating that both CFP-tagged proteins are functional. Both Ugt proteins co-localize with each other as well as with the GDP-mannose nucleotide transporter, as was demonstrated by fluorescence microscopy, thereby confirming their predicted localization in the Golgi. CONCLUSION: A. niger contains two genes encoding UDP-Galf-transporters. Deletion and localization studies indicate that UgtA and UgtB have redundant functions in the biosynthesis of Galf-containing glycoconjugates.


Asunto(s)
Aspergillus niger/metabolismo , Galactosa/análogos & derivados , Aparato de Golgi/metabolismo , Transferasas/metabolismo , Uridina Difosfato/análogos & derivados , Aspergillus niger/química , Aspergillus niger/genética , Pared Celular/metabolismo , Evolución Molecular , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Galactosa/metabolismo , Eliminación de Gen , Duplicación de Gen , Homología de Secuencia de Ácido Nucleico , Transferasas/química , Transferasas/genética , Uridina Difosfato/metabolismo
12.
FASEB J ; 28(1): 288-99, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24043262

RESUMEN

Atherosclerosis is a lipid-driven inflammatory disease of the vessel wall, characterized by the chronic activation of macrophages. We investigated whether the helminth-derived antigens [soluble egg antigens (SEAs)] could modulate macrophage inflammatory responses and protect against atherosclerosis in mice. In bone marrow-derived macrophages, SEAs induce anti-inflammatory macrophages, typified by high levels of IL-10 and reduced secretion of proinflammatory mediators. In hyperlipidemic LDLR(-/-) mice, SEA treatment reduced plaque size by 44%, and plaques were less advanced compared with PBS-injected littermate controls. The atheroprotective effect of SEAs was found to be mainly independent of cholesterol lowering and T-lymphocyte responses but instead could be attributed to diminished myeloid cell activation. SEAs reduced circulating neutrophils and inflammatory Ly6C(high) monocytes, and macrophages showed high IL-10 production. In line with the observed systemic effects, atherosclerotic lesions of SEA-treated mice showed reduced intraplaque inflammation as inflammatory markers [TNF-α, monocyte chemotactic protein 1 (MCP-1), intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and CD68], neutrophil content, and newly recruited macrophages were decreased. We show that SEA treatment protects against atherosclerosis development by dampening inflammatory responses. In the future, helminth-derived components may provide novel opportunities to treat chronic inflammatory diseases, as they diminish systemic inflammation and reduce the activation of immune cells.


Asunto(s)
Antígenos Helmínticos/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/terapia , Macrófagos/metabolismo , Animales , Quimiocina CCL2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Receptores de LDL/genética , Receptores de LDL/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Glycobiology ; 24(7): 602-18, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24727442

RESUMEN

Schistosomiasis is a debilitating parasitic disease of humans, endemic in tropical areas, for which no vaccine is available. Evidence points to glycan antigens as being important in immune responses to infection. Here we describe our studies on the comparative humoral immune responses to defined schistosome-type glycan epitopes in Schistosoma mansoni-infected humans, rhesus monkeys and mice. Rhesus anti-glycan responses over the course of infection were screened on a defined glycan microarray comprising semi-synthetic glycopeptides terminating with schistosome-associated or control mammalian-type glycan epitopes, as well as a defined glycan microarray of mammalian-type glycans representing over 400 glycan structures. Infected rhesus monkeys generated a high immunoglobulin G (IgG) antibody response to the core xylose/core α3 fucose epitope of N-glycans, which peaked at 8-11 weeks post infection, coinciding with maximal ability to kill schistosomula in vitro. By contrast, infected humans generated low antibody levels to this epitope. At 18 months following praziquantel therapy to eliminate the parasite, antibody levels were negligible. Mice chronically infected with S. mansoni generated high levels of anti-fucosylated LacdiNAc (GalNAcß1, 4(Fucα1, 3)GlcNAc) IgM antibodies, but lacked a robust response to the core xylose/core α3 fucose N-glycan antigens compared with other species studied, and their sera demonstrated an intermediate level of schistosomula killing in vitro. These differential responses to parasite glycan antigens may be related to the ability of rhesus monkeys to self-cure in contrast to the chronic infection seen in humans and mice. Our results validate defined glycan microarrays as a useful technology to evaluate diagnostic and vaccine antigens for schistosomiasis and perhaps other infections.


Asunto(s)
Anticuerpos Antihelmínticos/inmunología , Inmunoglobulina G/inmunología , Lactosa/análogos & derivados , Esquistosomiasis mansoni/inmunología , Adulto , Animales , Epítopos , Humanos , Lactosa/inmunología , Macaca mulatta , Ratones , Praziquantel/uso terapéutico , Esquistosomiasis mansoni/tratamiento farmacológico , Especificidad de la Especie
14.
J Biol Chem ; 287(49): 41523-33, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23035125

RESUMEN

Cosmc is the specific molecular chaperone in the endoplasmic reticulum for T-synthase, a Golgi ß3-galactosyltransferase that generates the core 1 O-glycan, Galß1-3GalNAcα-Ser/Thr, in glycoproteins. Dysfunctional Cosmc results in the formation of inactive T-synthase and consequent expression of the Tn antigen (GalNAcα1-Ser/Thr), which is associated with several human diseases. However, the molecular regulation of expression of Cosmc, which is encoded by a single gene on Xq24, is poorly understood. Here we show that epigenetic silencing of Cosmc through hypermethylation of its promoter leads to loss of Cosmc transcripts in Tn4 cells, an immortalized B cell line from a male patient with a Tn-syndrome-like phenotype. These cells lack T-synthase activity and express the Tn antigen. Treatment of cells with 5-aza-2'-deoxycytidine causes restoration of Cosmc transcripts, restores T-synthase activity, and reduces Tn antigen expression. Bisulfite sequencing shows that CG dinucleotides in the Cosmc core promoter are hypermethylated. Interestingly, several other X-linked genes associated with glycosylation are not silenced in Tn4 cells, and we observed no correlation of a particular DNA methyltransferase to aberrant methylation of Cosmc in these cells. Thus, hypermethylation of the Cosmc promoter in Tn4 cells is relatively specific. Epigenetic silencing of Cosmc provides another mechanism underlying the abnormal expression of the Tn antigen, which may be important in understanding aberrant Tn antigen expression in human diseases, including IgA nephropathy and cancer.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Epigénesis Genética , Regulación de la Expresión Génica , Leucocitos/metabolismo , Chaperonas Moleculares/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citosol/metabolismo , Silenciador del Gen , Glicosilación , Glicosiltransferasas/metabolismo , Humanos , Masculino , Metilación , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Transcripción Genética
15.
Glycobiology ; 22(5): 676-95, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22241826

RESUMEN

Immune responses induced by glycans upon infection with Schistosoma mansoni may be mediated by either schistosomal glycoproteins or glycosphingolipids. In this study, we have elucidated the structural features of both carbohydrate moieties and respective ceramide units of complex glycosphingolipids from adult S. mansoni. Obtained data revealed a vast structural heterogeneity due to manifold combinations of different oligosaccharides and ceramide entities. Observed carbohydrate moieties included Lewis(X) (Le(X); Gal(ß1-4)[Fuc(α1-3)]GlcNAc) as well as, in part, multiply fucosylated LacdiNAc (LDN; GalNAc(ß1-4)GlcNAc) carbohydrate epitopes. Corresponding lipid portions comprised predominantly C18-sphingosine as well as C18- and C20-phytosphingosine derivatives. Intriguingly, glycosphingolipids carrying an Le(X) epitope contained predominantly C18-sphingosine, whereas LDN-based species exhibited mostly phytosphingosine derivatives, in addition to C18-sphingosine, indicating that the two classes of glycosphingolipids might be synthesized via different biosynthetic routes. Compared with literature data, adult worm glycosphingolipids with Le(X) epitopes revealed clear structural differences in comparison to corresponding cercarial species which have been shown to exhibit mainly sphinganine bases with 18-21 carbon atoms. Therefore, it may be hypothesized that the divergent structural features of the respective ceramide moieties are responsible for the published observation that only adult worm, but not cercarial glycosphingolipids are able to induce dendritic cell activation skewing the T-cell response toward a Th1 profile.


Asunto(s)
Glicoesfingolípidos/metabolismo , Schistosoma mansoni/metabolismo , Animales , Conformación de Carbohidratos , Secuencia de Carbohidratos , Ensayo de Inmunoadsorción Enzimática , Glicoesfingolípidos/química , Datos de Secuencia Molecular , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
16.
Glycobiology ; 22(4): 456-69, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21940757

RESUMEN

Galactofuranose (Galf) is the five-membered ring form of galactose. It is widely distributed among several branches of the eukaryotic kingdom. This review highlights recent advances in our understanding of the biosynthesis and function of Galf-containing glycoconjugates in fungal Aspergillus spp. and the protozoan trypanosomatid parasites. We give an overview of the biosynthetic pathways leading to the production of glycolipids, glycoproteins and polysaccharides containing Galf in these species and their biological relevance. Remarkably, modification of the cell surface caused by Galf absence often results in morphological abnormalities and an impaired cell wall function in these organisms. Galf-deficient mutants are generally hypersensitive to drugs, exhibit a constitutive osmotic stress phenotype and/or have an attenuated virulence. Since Galf has never been found in mammals and higher plants, Galf-biosynthetic pathways have raised much interest as targets for drug development to combat microbial infections.


Asunto(s)
Galactosa/biosíntesis , Animales , Vías Biosintéticas , Conformación de Carbohidratos , Secuencia de Carbohidratos , Furanos/metabolismo , Galactosa/análogos & derivados , Galactosa/metabolismo , Galactosa/fisiología , Glucolípidos/biosíntesis , Glicoproteínas/biosíntesis , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Datos de Secuencia Molecular , Polisacáridos/biosíntesis , Polisacáridos/química , Polisacáridos/metabolismo , Estereoisomerismo
17.
Int J Cancer ; 131(1): 117-28, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21823122

RESUMEN

Altered glycosylation in epithelial cancers may play an important role in tumour progression, as it may affect tumour cell migration and antigen presentation by antigen presenting cells. We specifically characterise the glycosylation patterns of two tumour antigens that are highly expressed in cancer tissue and often detected in their secreted form in serum: the epithelial mucin MUC1 and carcinoembryonic antigen (CEA, also called CEACAM5). We analysed 48 colorectal cancer patients, comparing normal colon and tumour epithelium within each patient. Lectin binding was studied by a standardised CEA/MUC1 capture ELISA, using several plant lectins, and the human C-type lectins MGL and DC-SIGN, and Galectin-3. Peanut agglutinin (PNA) bound to MUC1 from tumour tissue in particular, suggests increased expression of the Thomsen-Friedenreich antigen (TF-antigen) (Core 1, Galß1-3GalNAc-Ser/Thr). Only small amounts of Tn-antigen (GalNAcα-Ser/Thr) expression was observed, but the human C-type lectin MGL showed increased binding to tumour-associated MUC1. Furthermore, sialylation was greatly enhanced. In sharp contrast, tumour-associated CEA (CEACAM5) contained high levels of the blood-group related carbohydrates, Lewis X and Lewis Y. This correlated strongly with the interaction of the human C-type lectin DC-SIGN to tumour-associated CEA, suggesting that CEA can be recognized and taken up by antigen presenting cells. In addition, increased mannose expression was observed and branched N-glycans were prominent, and this correlated well with human Galectin-3 binding. These data demonstrate that individual tumour antigens contain distinct glycan structures associated with cancer and, since glycans affect cellular interactions with its microenvironment, this may have consequences for progression of the disease.


Asunto(s)
Antígeno Carcinoembrionario/metabolismo , Colon/metabolismo , Neoplasias del Colon/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Intestinal/metabolismo , Mucina-1/metabolismo , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neoplasias del Colon/patología , Proteínas Ligadas a GPI/metabolismo , Galectina 3/metabolismo , Glicosilación , Humanos , Lectinas Tipo C/metabolismo , Antígenos del Grupo Sanguíneo de Lewis/biosíntesis , Antígeno Lewis X/biosíntesis , Manosa/biosíntesis , Aglutinina de Mani/metabolismo , Receptores de Superficie Celular/metabolismo
18.
Biol Chem ; 393(8): 767-75, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22944679

RESUMEN

Aspergillus niger possesses a galactofuranosidase activity, however, the corresponding enzyme or gene encoding this enzyme has never been identified. As evidence is mounting that enzymes exist with affinity for both arabinofuranose and galactofuranose, we investigated the possibility that α-L-arabinofuranosidases, encoded by the abfA and abfB genes, are responsible for the galactofuranosidase activity of A. niger. Characterization of the recombinant AbfA and AbfB proteins revealed that both enzymes do not only hydrolyze p-nitrophenyl-α-L-arabinofuranoside (pNp-α-Araf) but are also capable of hydrolyzing p-nitrophenyl-ß-D-galactofuranoside (pNp-ß-Galf). Molecular modeling of the AbfB protein with pNp-ß-Galf confirmed the possibility for AbfB to interact with this substrate, similarly as with pNp-α-Araf. We also show that galactomannan, a cell wall compound of A. niger, containing ß-linked terminal and internal galactofuranosyl moieties, can be degraded by an enzyme activity that is present in the supernatant of inulin-grown A. niger. Interestingly, purified AbfA and AbfB did not show this hydrolyzing activity toward A. nigergalactomannan. In summary, our studies demonstrate that AbfA and AbfB, α-L-arabinofuranosidases from different families, both contain a galactofuranose (Galf)-hydrolyzing activity. In addition, our data support the presence of a Galf-hydrolase activity expressed by A. niger that is capable of degrading fungal galactomannan.


Asunto(s)
Aspergillus niger/enzimología , Galactosidasas/metabolismo , Glicósido Hidrolasas/metabolismo , Arabinosa/análogos & derivados , Arabinosa/metabolismo , Aspergillus niger/metabolismo , Galactosa/análogos & derivados , Galactósidos/metabolismo , Glucosa/metabolismo , Hidrólisis , Inulina/metabolismo , Mananos/metabolismo , Modelos Moleculares , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
19.
Sci Adv ; 8(43): eabm8783, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36306365

RESUMEN

The underlying pathology of immunoglobulin A (IgA) nephropathy (IgAN), the most common glomerulonephritis worldwide, is driven by the deposition of immune complexes containing galactose-deficient IgA1 [Tn(+)IgA1] in the glomerular mesangium. Here, we report that novel anti-Tn circulating immune complexes (anti-Tn CICs) contain predominantly IgM, representing large macromolecular complexes of ~1.2 megadaltons to several megadalton sizes together with Tn(+)IgA1 and some IgG. These complexes are significantly elevated in sera of patients with IgAN, which contains higher levels of complement C3, compared to healthy individuals. Anti-Tn CICs are bioactive and induce specific proliferation of human renal mesangial cells. We found that these anti-Tn CICs can be dissociated with small glycomimetic compounds, which mimic the Tn antigen of Tn(+)IgA1, releasing IgA1 from anti-Tn CICs. This glycomimetic compound can also significantly inhibit the proliferative activity of anti-Tn CICs of patients with IgAN. These findings could enhance both the diagnosis of IgAN and its treatment, as specific drug treatments are now unavailable.


Asunto(s)
Glomerulonefritis por IGA , Humanos , Glomerulonefritis por IGA/tratamiento farmacológico , Complejo Antígeno-Anticuerpo , Mesangio Glomerular , Inmunoglobulina A , Células Mesangiales
20.
Exp Parasitol ; 129(3): 221-6, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21893057

RESUMEN

Diagnostic methods for parasite infections still highly depend on the identification of the parasites by direct methods such as microscopic examination of blood, stool and tissue biopsies. Serodiagnosis is often carried out to complement the direct methods; however, few synthetic antigens with sufficient sensitivity and specificity are available. Here we evaluated a glycan microarray approach to select for synthetic glycan antigens that could be used for serodiagnosis of parasitic infections. Using a glycan array containing over 250 different glycan antigens, we identified GalNAcß1-4(Fucα1-3)GlcNAc-R (LDNF) as a glycan antigen that is recognized by antibodies from Trichinella-infected individuals. We synthesized a neoglycoconjugate, consisting of five LDNF molecules covalently coupled to bovine serum albumin (BSA), and used this neoglycoconjugate as an antigen to develop a highly sensitive total-Ig ELISA for serological screening of trichinellosis. The results indicate that glycan microarrays constitute a promising technology for fast and specific identification of parasite glycan antigens to improve serodiagnosis of different parasitic infections, either using an ELISA format, or parasite-specific glycan arrays.


Asunto(s)
Anticuerpos Antihelmínticos/sangre , Lactosa/análogos & derivados , Polisacáridos/inmunología , Trichinella spiralis/inmunología , Triquinelosis/diagnóstico , Animales , Anticuerpos Antihelmínticos/inmunología , Anticuerpos Monoclonales/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Lactosa/inmunología , Masculino , Análisis por Matrices de Proteínas , Ratas , Ratas Wistar , Sensibilidad y Especificidad , Triquinelosis/sangre , Triquinelosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA