Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Int Immunopharmacol ; 137: 112512, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38897123

RESUMEN

OBJECTIVE: This study aims to disclose how loss of fucosyltransferase 2 (Fut2) impacts intestinal inflammation through cGAS-STING pathway that is closely associated with gut microbiota, and which microbial metabolite improves colitis in Fut2 deficiency. METHODS: Chronic colitis was induced in intestinal epithelial Fut2 knock out mice (Fut2△IEC), whose intestinal inflammation and activity of cGAS-STING pathway were evaluated. 16S rRNA sequencing and metabolomics were performed using intestinal samples. 2-oxindole was used to treat RAW264.7 cells and Fut2△IEC mice with colitis (Fut2△IEC-DSS) to investigate the effect of 2-oxindole on cGAS-STING response and intestinal inflammation. RESULTS: Fut2 loss exacerbated chronic colitis in mice, manifested by declined body weight, reduced colon length, increased disease activity index (DAI) and more colon injury in Fut2△IEC-DSS mice compared with WT-DSS (wild type mice with colitis). Lack of Fut2 promoted activation of cGAS-STING pathway. Fut2 deficiency had a primary impact on colonic microbiota, as shown by alteration of microbial diversity and structure, as well as decreased Lactobacillus. Metabolic structure and tryptophan metabolism in colonic luminal microbiota were also influenced by Fut2 loss. Fut2 deficiency also led to decreased levels of aryl hydrocarbon receptor (AHR) and its ligand 2-oxindole derived from tryptophan metabolism. 2-oxindole compromised cGAS-STING response through activating AHR in macrophages, and protected against intestinal inflammation and overactive cGAS-STING pathway in Fut2△IEC-DSS mice. CONCLUSION: Fut2 deficiency promotes cGAS-STING pathway through suppressing 2-oxindole-AHR axis, ultimately facilitating the susceptibility to chronic colitis.


Asunto(s)
Colitis , Fucosiltransferasas , Microbioma Gastrointestinal , Proteínas de la Membrana , Ratones Noqueados , Nucleotidiltransferasas , Oxindoles , Transducción de Señal , Animales , Humanos , Masculino , Ratones , Enfermedad Crónica , Colitis/inducido químicamente , Colitis/inmunología , Colon/patología , Colon/inmunología , Colon/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Fucosiltransferasas/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Células RAW 264.7 , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hidrocarburo de Aril/genética
2.
Immunohorizons ; 5(8): 703-710, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433625

RESUMEN

Allergic skin inflammation requires the influx of inflammatory cells into the skin. Extravasation of leukocytes into the skin requires interactions between endothelial selectins and their glycan ligands on the surface of leukocytes. Selectin-ligand formation requires the activity of several glycosyltransferases, including Fut7 In this report, we tested the importance of Fut7 for the development of allergic skin inflammation in the Stat6VT transgenic mouse model. We observed that Fut7 deficiency was protective but did not eliminate disease. Segregation of the data by gender of the parent that transmitted the Stat6VT transgene, but not by gender of the pups, which were analyzed for disease, revealed that the protective effects of Fut7 deficiency were significantly greater when dams were Stat6VT negative. In contrast, in mice from litters of Stat6VT+ dams, Fut7 deficiency resulted in only modest protection. These findings indicate that pups from atopic dams exhibit a greater propensity for allergic disease, similar to observations in humans, and that the effect of maternal atopy is due to enhanced selectin-independent mechanisms of leukocyte recruitment in their offspring. Together, these results demonstrate that Fut7 deficiency can be protective in a model of atopic dermatitis but that maternal atopy diminishes these protective effects, suggesting alternative pathways for leukocyte recruitment in the absence of Fut7 enzyme activity. These observations have implications for understanding how the environment in utero predisposes for the development of allergic disease.


Asunto(s)
Dermatitis Atópica/inmunología , Selectina E/inmunología , Inmunidad Materno-Adquirida/inmunología , Inflamación/inmunología , Selectina-P/inmunología , Piel/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Dermatitis Atópica/genética , Dermatitis Atópica/metabolismo , Selectina E/metabolismo , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Fucosiltransferasas/inmunología , Humanos , Inmunidad Materno-Adquirida/genética , Inflamación/genética , Inflamación/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Selectina-P/metabolismo , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/inmunología , Factor de Transcripción STAT6/metabolismo , Piel/metabolismo , Piel/patología
3.
Commun Biol ; 4(1): 832, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215830

RESUMEN

Sialyl-Lewis x (sLex, CD15s) is a tetra-saccharide on the surface of leukocytes required for E-selectin-mediated rolling, a prerequisite for leukocytes to migrate out of the blood vessels. Here we show using flow cytometry that sLex expression on basophils and mast cell progenitors depends on fucosyltransferase 6 (FUT6). Using genetic association data analysis and qPCR, the cell type-specific defect was associated with single nucleotide polymorphisms (SNPs) in the FUT6 gene region (tagged by rs17855739 and rs778798), affecting coding sequence and/or expression level of the mRNA. Heterozygous individuals with one functional FUT6 gene harbor a mixed population of sLex+ and sLex- basophils, a phenomenon caused by random monoallelic expression (RME). Microfluidic assay demonstrated FUT6-deficient basophils rolling on E-selectin is severely impaired. FUT6 null alleles carriers exhibit elevated blood basophil counts and a reduced itch sensitivity against insect bites. FUT6-deficiency thus dampens the basophil-mediated allergic response in the periphery, evident also in lower IgE titers and reduced eosinophil counts.


Asunto(s)
Basófilos/metabolismo , Fucosiltransferasas/genética , Expresión Génica , Antígeno Sialil Lewis X/biosíntesis , Secuencia de Bases , Basófilos/citología , Células Cultivadas , Estudios de Cohortes , Selectina E/metabolismo , Fucosiltransferasas/deficiencia , Perfilación de la Expresión Génica/métodos , Humanos , Recuento de Leucocitos , Rodamiento de Leucocito/genética , Rodamiento de Leucocito/fisiología , Polimorfismo de Nucleótido Simple , Homología de Secuencia de Ácido Nucleico
4.
J Biomed Sci ; 28(1): 20, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33722220

RESUMEN

BACKGROUND AND AIMS: Previous study disclosed Fucosyltransferase 2 (Fut2) gene as a IBD risk locus. This study aimed to explore the mechanism of Fut2 in IBD susceptibility and to propose a new strategy for the treatment of IBD. METHODS: Intestinal epithelium-specific Fut2 knockout (Fut2△IEC) mice was used. Colitis was induced by dextran sulfate sodium (DSS). The composition and diversity of gut microbiota were assessed via 16S rRNA analysis and the metabolomic findings was obtained from mice feces via metabolite profiling. The fecal microbiota transplantation (FMT) experiment was performed to confirm the association of gut microbiota and LPC. WT mice were treated with Lysophosphatidylcholine (LPC) to verify its impact on colitis. RESULTS: The expression of Fut2 and α-1,2-fucosylation in colonic tissues were decreased in patients with UC (UC vs. control, P = 0.036) and CD (CD vs. control, P = 0.031). When treated with DSS, in comparison to WT mice, more severe intestinal inflammation and destructive barrier functions in Fut2△IEC mice was noted. Lower gut microbiota diversity was observed in Fut2△IEC mice compared with WT mice (p < 0.001). When exposed to DSS, gut bacterial diversity and composition altered obviously in Fut2△IEC mice and the fecal concentration of LPC was increased. FMT experiment revealed that mice received the fecal microbiota from Fut2△IEC mice exhibited more severe colitis and higher fecal LPC concentration. Correlation analysis showed that the concentration of LPC was positively correlated with four bacteria-Escherichia, Bilophila, Enterorhabdus and Gordonibacter. Furthermore, LPC was proved to promote the release of pro-inflammatory cytokines and damage epithelial barrier in vitro and in vivo. CONCLUSION: Fut2 and α-1,2-fucosylation in colon were decreased not only in CD but also in UC patients. Gut microbiota in Fut2△IEC mice is altered structurally and functionally, promoting generation of LPC which was proved to promote inflammation and damage epithelial barrier.


Asunto(s)
Bacterias/metabolismo , Colitis/microbiología , Fucosiltransferasas/deficiencia , Microbioma Gastrointestinal , Lisofosfatidilcolinas/metabolismo , Animales , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Transgénicos , Galactósido 2-alfa-L-Fucosiltransferasa
5.
Cell Mol Gastroenterol Hepatol ; 12(1): 293-320, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33631374

RESUMEN

BACKGROUND & AIMS: Fucosyltransferase 2 (Fut2)-mediated intestinal α1- 2-fucosylation is important for host-microbe interactions and has been associated with several diseases, but its role in obesity and hepatic steatohepatitis is not known. The aim of this study was to investigate the role of Fut2 in a Western-style diet-induced mouse model of obesity and steatohepatitis. METHODS: Wild-type (WT) and Fut2-deficient littermate mice were used and features of the metabolic syndrome and steatohepatitis were assessed after 20 weeks of Western diet feeding. RESULTS: Intestinal α1-2-fucosylation was suppressed in WT mice after Western diet feeding, and supplementation of α1-2-fucosylated glycans exacerbated obesity and steatohepatitis in these mice. Fut2-deficient mice were protected from Western diet-induced features of obesity and steatohepatitis despite an increased caloric intake. These mice have increased energy expenditure and thermogenesis, as evidenced by a higher core body temperature. Protection from obesity and steatohepatitis associated with Fut2 deficiency is transmissible to WT mice via microbiota exchange; phenotypic differences between Western diet-fed WT and Fut2-deficient mice were reduced with antibiotic treatment. Fut2 deficiency attenuated diet-induced bile acid accumulation by altered relative abundance of bacterial enzyme 7-α-hydroxysteroid dehydrogenases metabolizing bile acids and by increased fecal excretion of secondary bile acids. This also was associated with increased intestinal farnesoid X receptor/fibroblast growth factor 15 signaling, which inhibits hepatic synthesis of bile acids. Dietary supplementation of α1-2-fucosylated glycans abrogates the protective effects of Fut2 deficiency. CONCLUSIONS: α1-2-fucosylation is an important host-derived regulator of intestinal microbiota and plays an important role for the pathogenesis of obesity and steatohepatitis in mice.


Asunto(s)
Hígado Graso/metabolismo , Fucosiltransferasas/metabolismo , Intestinos/enzimología , Obesidad/metabolismo , Animales , Dieta , Hígado Graso/inducido químicamente , Fucosiltransferasas/deficiencia , Intestinos/microbiología , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Galactósido 2-alfa-L-Fucosiltransferasa
6.
J Biol Chem ; 295(46): 15742-15753, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32913123

RESUMEN

ADAMTSL2 mutations cause an autosomal recessive connective tissue disorder, geleophysic dysplasia 1 (GPHYSD1), which is characterized by short stature, small hands and feet, and cardiac defects. ADAMTSL2 is a matricellular protein previously shown to interact with latent transforming growth factor-ß binding protein 1 and influence assembly of fibrillin 1 microfibrils. ADAMTSL2 contains seven thrombospondin type-1 repeats (TSRs), six of which contain the consensus sequence for O-fucosylation by protein O-fucosyltransferase 2 (POFUT2). O-fucose-modified TSRs are subsequently elongated to a glucose ß1-3-fucose (GlcFuc) disaccharide by ß1,3-glucosyltransferase (B3GLCT). B3GLCT mutations cause Peters Plus Syndrome (PTRPLS), which is characterized by skeletal defects similar to GPHYSD1. Several ADAMTSL2 TSRs also have consensus sequences for C-mannosylation. Six reported GPHYSD1 mutations occur within the TSRs and two lie near O-fucosylation sites. To investigate the effects of TSR glycosylation on ADAMTSL2 function, we used MS to identify glycan modifications at predicted consensus sequences on mouse ADAMTSL2. We found that most TSRs were modified with the GlcFuc disaccharide at high stoichiometry at O-fucosylation sites and variable mannose stoichiometry at C-mannosylation sites. Loss of ADAMTSL2 secretion in POFUT2-/- but not in B3GLCT-/- cells suggested that impaired ADAMTSL2 secretion is not responsible for skeletal defects in PTRPLS patients. In contrast, secretion was significantly reduced for ADAMTSL2 carrying GPHYSD1 mutations (S641L in TSR3 and G817R in TSR6), and S641L eliminated O-fucosylation of TSR3. These results provide evidence that abnormalities in GPHYSD1 patients with this mutation are caused by loss of O-fucosylation on TSR3 and impaired ADAMTSL2 secretion.


Asunto(s)
Proteínas ADAMTS/metabolismo , Enfermedades del Desarrollo Óseo/patología , Proteínas de la Matriz Extracelular/metabolismo , Deformidades Congénitas de las Extremidades/patología , Proteínas ADAMTS/química , Proteínas ADAMTS/genética , Secuencia de Aminoácidos , Animales , Enfermedades del Desarrollo Óseo/genética , Sistemas CRISPR-Cas/genética , Disacáridos/química , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Edición Génica , Glicosilación , Glicosiltransferasas/deficiencia , Glicosiltransferasas/genética , Células HEK293 , Humanos , Deformidades Congénitas de las Extremidades/genética , Manosa/química , Ratones , Mutagénesis Sitio-Dirigida , Dominios Proteicos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia
7.
Sci Rep ; 10(1): 14178, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32843681

RESUMEN

Systemic lupus erythematosus (SLE) is a complex multi-system autoimmune disease. Detection of anti-nuclear antibodies (ANA) is fundamental for the diagnosis of SLE. In the present study, we found that the level of core fucosylation catalyzed by α1,6-fucosyltransferase (Fut8) is markedly up-regulated on immunoglobulin G (IgG) in the sera of SLE patients detected by Aspergillus oryzae lectin (AOL) blot. In sandwich Dot enzyme-linked immunosorbent assay (Dot-ELISA), the core fucosylation level was also found significantly increased in the sera from SLE patients with a higher ANA titer. To establish a rapid and sensitive laboratory test for the diagnosis of SLE, we prokaryotically expressed AOL and C3-D1-C3-D2-C3 of protein G (SpG3), and generate AOL-conjugated colloid gold immunochromatographic strips (ICS). The detection limit of core fucosylated IgG was 10 µg/mL for AOL-conjugated colloid gold ICS. As well as indirect immunofluorescence, the AOL-conjugated colloid gold ICS showed reliable results in the serum of 39 SLE patients. Our results indicated that the AOL-conjugated colloid gold ICS could serve as a rapid test for the detection of ANA and suspected cases of SLE.


Asunto(s)
Cromatografía de Afinidad , Inmunoglobulina G/química , Lupus Eritematoso Sistémico/diagnóstico , Tiras Reactivas , Adolescente , Adulto , Anciano , Animales , Anticuerpos Antinucleares/sangre , Aspergillus oryzae , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biomarcadores , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Fucosa/metabolismo , Fucosiltransferasas/deficiencia , Fucosiltransferasas/metabolismo , Glicosilación , Oro Coloide , Humanos , Immunoblotting , Inmunoglobulina G/sangre , Lupus Eritematoso Sistémico/sangre , Masculino , Ratones Noqueados , Persona de Mediana Edad , Lectinas de Plantas , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión , Organismos Libres de Patógenos Específicos , Adulto Joven
8.
Cell Death Dis ; 11(4): 285, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32332708

RESUMEN

Fucosylation is a biological process that plays a critical role in multiple cellular functions from cell adhesion to immune regulation. Fucosyltransferases (FUTs) mediate fucosylation, and dysregulation of genes encoding FUTs is associated with various diseases. FUT1 and its fucosylated products are expressed in the ocular surface and ocular adnexa; however, the role of FUT1 in the ocular surface health and disease is yet unclear. Here, we investigated the effects of FUT1 on the ocular surface in steady-state conditions with age and under desiccating stress using a Fut1 knockout (KO) mouse model. We found that corneal epithelial defects and stromal opacity developed in Fut1 KO mice. Also, inflammatory responses in the ocular surface and Th1 cell activation in ocular draining lymph nodes (DLNs) were upregulated. Desiccating stress further aggravated Th1 cell-mediated immune responses in DLNs, lacrimal gland, and ocular surface in Fut1 KO mice, leading to severe corneal epithelial disruption and opacity. Mixed lymphocyte reaction assays revealed that the activity of splenocytes to stimulate CD4 T-cell proliferation was increased in Fut1 KO mice. Together, these data demonstrate that FUT1 deficiency induces immune dysregulation in the ocular surface and corneal opacity in steady state and under desiccating stress.


Asunto(s)
Opacidad de la Córnea/inducido químicamente , Fucosiltransferasas/deficiencia , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Galactósido 2-alfa-L-Fucosiltransferasa
9.
Biochem J ; 477(6): 1179-1201, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32141499

RESUMEN

Fucosyltransferase 8 (FUT8) and ß-galactoside α-2,6-sialyltransferase 1 (ST6GAL1) are glycosyltransferases that catalyze α1,6-fucosylation and α2,6-sialylation, respectively, in the mammalian N-glycosylation pathway. They are aberrantly expressed in various human diseases. FUT8 is non-glycosylated but is responsible for the fucosylation of ST6GAL1. However, the mechanism for the interaction between these two enzymes is unknown. In this study, we show that serum levels of α2,6-sialylated N-glycans are increased in Fut8-/- mice, whereas the mRNA and protein levels of ST6GAL1 are unchanged in mouse live tissues. The level of α2,6-sialylation on IgG was also enhanced in Fut8-/- mice along with ST6GAL1 catalytic activity increase in both serum and liver. Moreover, it was observed that ST6GAL1 prefers non-fucosylated substrates. Interestingly, increased core fucosylation accompanied by a reduction in α2,6-sialylation, was detected in rheumatoid arthritis patient serum. These findings provide new insight into the interactions between FUT8 and ST6GAL1.


Asunto(s)
Antígenos CD/genética , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Sialiltransferasas/deficiencia , Sialiltransferasas/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Femenino , Fucosa/genética , Fucosa/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilación , Humanos , Ratones , Ratones Noqueados , Persona de Mediana Edad
10.
J Inherit Metab Dis ; 43(4): 871-879, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32049367

RESUMEN

Pathogenic variants in the Golgi localised alpha 1,6 fucosyltransferase, FUT8, cause a rare inherited metabolic disorder known as FUT8-CDG. To date, only three affected individuals have been reported presenting with a constellation of symptoms including intrauterine growth restriction, severe delays in growth and development, other neurological impairments, significantly shortened limbs, respiratory complications, and shortened lifespan. Here, we report an additional four unrelated affected individuals homozygous for novel pathogenic variants in FUT8. Analysis of serum N-glycans revealed a complete lack of core fucosylation, an important diagnostic biomarker of FUT8-CDG. Our data expands both the molecular and clinical phenotypes of FUT8-CDG and highlights the importance of identifying a reliable biomarker for confirming potentially pathogenic variants.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Fucosa/metabolismo , Fucosiltransferasas/genética , Polisacáridos/metabolismo , Trastornos Congénitos de Glicosilación/metabolismo , Femenino , Fucosiltransferasas/deficiencia , Humanos , Masculino , Espectrometría de Masas , Fenotipo , Secuenciación del Exoma
11.
Biochem Biophys Res Commun ; 520(3): 612-618, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31623829

RESUMEN

BACKGROUND: FUT8-mediated core fucosylation, which transfers a fucose residue from GDP-fucose to core-GlcNAc of the N-linked type glycoproteins, is crucial for signaling receptors function. Core fucosylation is involved in various biological processes such as cell proliferation, apoptosis, differentiation and immune regulation. Our previous studies demonstrated that inhibiting core fucosylation prevented renal interstitial fibrosis of UUO murine models, but its role in the development of diabetic kidney disease (DKD) remains unclear. This study aimed to clarify the protective effects and molecular mechanisms during the progress of DKD by inhibiting core fucosylation in vivo. METHODS: Core fucosylation was examined in streptozotocin (STZ)-induced diabetic mouse model. Then a new Fut8 mutation mouse model in which exon 7 of Fut8 gene is deleted was constructed for diabetes induction. Metabolic and renal parameters were measured. Renal structure, fibrosis, and podocyte injury were assessed, and underlying mechanisms were investigated. RESULTS: The levels of fasting blood glucose, glycated hemoglobin, kidney-weight-to- body-weight (KW/BW) and urine albumin-to-creatinine (ACR) were increased at 16 weeks post injection. KW/BW and urine ACR were decreased significantly by inhibiting core fucosylation. The renal pathology, fibrosis, and podocyte injury were mitigated significantly by inhibiting core fucosylation. The protective effects of inhibiting core fucosylation were mediated by downregulated of the phosphorylation of Smad2/3 and extracellular signal-regulated kinase (ERK). CONCLUSIONS: Our results indicate that FUT8-based treatment might be a promising intervention strategy in therapeutic paradigm of DKD.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Fucosa/metabolismo , Animales , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/patología , Progresión de la Enfermedad , Regulación hacia Abajo , Fibrosis , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Glicosilación , Riñón/metabolismo , Riñón/patología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Transducción de Señal , Proteínas Smad/metabolismo
12.
Anal Sci ; 35(12): 1333-1340, 2019 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-31423004

RESUMEN

Human antithrombin (AT) has two isoforms of which the predominant α-form is glycosylated on all four possible glycosylation sites and the lower abundant ß-isoform lacks the oligosaccharide on Asn135. The main oligosaccharide structure of human AT consists of biantennary complex-type oligosaccharides lacking a core fucose. Generally, Chinese hamster ovary (CHO) cells produce recombinant human AT (rhAT) with core-fucosylated oligosaccharides. However, rhAT lacking core-fucose oligosaccharides can be produced by POTELLIGENT® technology, which uses FUT8 knockout CHO cells in production. The rhAT has more variable glycan structures, such as tetra-antennary complex type, high-mannose type, and mannose 6-phosphate species as minor components compared to plasma-derived human AT (phAT). In addition, the site-specific glycan profile was different between two ATs. We evaluated the effect of these properties on efficacy and safety based on a comparison of rhAT made by that technology with phAT in terms of their respective oligosaccharide structures, site-specific oligosaccharide profiles, and the ratio of α- and ß-forms. Although some structural differences were found between the rhAT and phAT, we concluded that these differences have no significant effect on the efficacy and safety of rhAT.


Asunto(s)
Antitrombinas/química , Antitrombinas/metabolismo , Ingeniería Genética/métodos , Oligosacáridos/química , Plasma/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Técnicas de Inactivación de Genes , Glicosilación , Humanos , Proteínas Recombinantes/genética
13.
PLoS Pathog ; 15(7): e1007915, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31329635

RESUMEN

Expression of ABO and Lewis histo-blood group antigens by the gastrointestinal epithelium is governed by an α-1,2-fucosyltransferase enzyme encoded by the Fut2 gene. Alterations in mucin glycosylation have been associated with susceptibility to various bacterial and viral infections. Salmonella enterica serovar Typhimurium is a food-borne pathogen and a major cause of gastroenteritis. In order to determine the role of Fut2-dependent glycans in Salmonella-triggered intestinal inflammation, Fut2+/+ and Fut2-/- mice were orally infected with S. Typhimurium and bacterial colonization and intestinal inflammation were analyzed. Bacterial load in the intestine of Fut2-/- mice was significantly lower compared to Fut2+/+ mice. Analysis of histopathological changes revealed significantly lower levels of intestinal inflammation in Fut2-/- mice compared to Fut2+/+ mice and measurement of lipocalin-2 level in feces corroborated histopathological findings. Salmonella express fimbriae that assist in adherence of bacteria to host cells thereby facilitating their invasion. The std fimbrial operon of S. Typhimurium encodes the π-class Std fimbriae which bind terminal α(1,2)-fucose residues. An isogenic mutant of S. Typhimurium lacking Std fimbriae colonized Fut2+/+ and Fut2-/- mice to similar levels and resulted in similar intestinal inflammation. In vitro adhesion assays revealed that bacteria possessing Std fimbriae adhered significantly more to fucosylated cell lines or primary epithelial cells in comparison to cells lacking α(1,2)-fucose. Overall, these results indicate that Salmonella-triggered intestinal inflammation and colonization are dependent on Std-fucose interaction.


Asunto(s)
Fimbrias Bacterianas/metabolismo , Fucosa/metabolismo , Salmonella typhimurium/patogenicidad , Animales , Adhesión Bacteriana , Colitis/etiología , Colitis/metabolismo , Colitis/microbiología , Femenino , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/genética , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Interacciones Microbiota-Huesped , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos CBA , Ratones Noqueados , Operón , Salmonelosis Animal/etiología , Salmonelosis Animal/metabolismo , Salmonelosis Animal/microbiología , Salmonella typhimurium/genética , Salmonella typhimurium/fisiología , Galactósido 2-alfa-L-Fucosiltransferasa
14.
Cancer Biomark ; 25(4): 303-311, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31306109

RESUMEN

BACKGROUND AND OBJECTIVE: Fucosyltranferase 8 (FUT8), which catalyzes core fucosylation of glycopeptides, plays important roles in cancer development. In this study, we aimed to explore the influence of FUT8 expression on migration ability of human breast cancer cells and its potential mechanisms. METHODS: The core fucosylation levels in normal and FUT8 deficient MCF-7 cells were analyzed by lectin LCA blots. Then, the cell adhesion assay, transwell and wound healing experiments were conducted. The phosphorylation of FAK and core fucosylation of E-cadherin and its downstream integrins in the FAK/integrin pathway were measured. Moreover, the expression levels of nuclear ß-catenin, MMP-2, and MMP-9 were also measured. RESULTS: The core fucosylation levels were significantly reduced by inhibited FUT8. FUT8 deficiency suppressed the adhesion, migration and invasion of MCF-7 cells; the potential mechanisms might involve three aspects. FUT8 deficiency inhibited FAK/integrin pathway by suppressing core fucosylation of E-cadherin. In addition, FUT8 deficiency reduced nuclear ß-catenin accumulation. The suppression of MMP-2 and MMP-9 expression also accounted for FUT8 deficiency inhibiting breast cancer cells migration. CONCLUSIONS: FUT8 deficiency suppressed migration of MCF-7 cells by impacting core fucosylation of E-cadherin and the downstream FAK/integrin pathway. Therefore, FUT8 is a potential biomarker for breast cancer detection and treatment.


Asunto(s)
Neoplasias de la Mama/genética , Fucosiltransferasas/deficiencia , Integrinas/genética , Neoplasias de la Mama/patología , Movimiento Celular , Femenino , Humanos
15.
Bioprocess Biosyst Eng ; 42(8): 1263-1271, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30982137

RESUMEN

In recent years, an increasing number of defucosylated therapeutic antibodies have been applied in clinical practices due to their better efficacy compared to fucosylated counterparts. The establishment of stable and clonal manufacturing cell lines is the basis of therapeutic antibodies production. Bioprocess development of a new cell line is necessary for its future applications in the biopharmaceutical industry. We engineered a stable cell line expressing defucosylated anti-HER2 antibody based on an established α-1,6-fucosyltransferase (FUT8) gene knockout CHO-S cell line. The optimization of medium and feed was evaluated in a small-scale culture system. Then the optimal medium and feed were scaled up in a bioreactor system. After fed-batch culture over 13 days, we evaluated the cell growth, antibody yield, glycan compositions and bioactivities. The production of anti-HER2 antibody from the FUT8 gene knockout CHO-S cells in the bioreactor increased by 37% compared to the shake flask system. The N-glycan profile of the produced antibody was consistent between the bioreactor and shake flask system. The antibody-dependent cellular cytotoxicity activity of the defucosylated antibody increased 14-fold compared to the wild-type antibody, which was the same as our previous results. The results of our bioprocess development demonstrated that the engineered cell line could be developed to a biopharmaceutical industrial cell line.


Asunto(s)
Antineoplásicos Inmunológicos/metabolismo , Fucosiltransferasas/deficiencia , Eliminación de Gen , Receptor ErbB-2/antagonistas & inhibidores , Animales , Células CHO , Cricetulus , Glicosilación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética
16.
PLoS One ; 13(6): e0199128, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29924834

RESUMEN

The fucose salvage pathway is a two-step process in which mammalian cells transform L-fucose into GDP-L-fucose, a universal fucose donor used by fucosyltransferases to modify glycans. Emerging evidence indicates the fucose salvage pathway and the fucosylation of proteins are altered during melanoma progression and metastasis. However the underlying mechanisms are not completely understood. Here, we report that the fucose salvage pathway inhibits invadopodia formation and extracellular matrix degradation by promoting α-1,2 fucosylation. Chemically or genetically increasing the fucose salvage pathway decreases invadopodium numbers and inhibits the proteolytic activity of invadopodia in WM793 melanoma cells. Inhibiting fucosylation by depleting fucokinase abrogates L-fucose-mediated inhibition of invadopodia, suggesting dependence on the fucose salvage pathway. The inhibition of invadopodium formation by L-fucose or ectopically expressed FUK could be rescued by treatment with α-1,2, but not α-1,3/α-1,4 fucosidase, implicating an α-1,2 fucose linkage-dependent anti-metastatic effect. The expression of FUT1, an α-1,2 fucosyltransferase, is remarkably down-regulated during melanoma progression, and the ectopic expression of FUT1 is sufficient to inhibit invadopodium formation and ECM degradation. Our findings indicate that the fucose salvage pathway can inhibit invadopodium formation, and consequently, invasiveness in melanoma via α-1,2 fucosylation. Re-activation of this pathway in melanoma could be useful for preventing melanoma invasion and metastasis.


Asunto(s)
Matriz Extracelular/metabolismo , Fucosa/metabolismo , Fucosiltransferasas/fisiología , Melanoma/metabolismo , Proteínas de Neoplasias/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Podosomas/fisiología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Fucosa/farmacología , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Vectores Genéticos/farmacología , Glicosilación , Humanos , Melanoma/fisiopatología , Redes y Vías Metabólicas , Invasividad Neoplásica , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Podosomas/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/farmacología , Regulación hacia Arriba , Galactósido 2-alfa-L-Fucosiltransferasa
17.
Nat Commun ; 8(1): 578, 2017 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-28924218

RESUMEN

Coronary artery anomalies may cause life-threatening cardiac complications; however, developmental mechanisms underpinning coronary artery formation remain ill-defined. Here we identify an angiogenic cell population for coronary artery formation in mice. Regulated by a DLL4/NOTCH1/VEGFA/VEGFR2 signaling axis, these angiogenic cells generate mature coronary arteries. The NOTCH modulator POFUT1 critically regulates this signaling axis. POFUT1 inactivation disrupts signaling events and results in excessive angiogenic cell proliferation and plexus formation, leading to anomalous coronary arteries, myocardial infarction and heart failure. Simultaneous VEGFR2 inactivation fully rescues these defects. These findings show that dysregulated angiogenic precursors link coronary anomalies to ischemic heart disease.Though coronary arteries are crucial for heart function, the mechanisms guiding their formation are largely unknown. Here, Wang et al. identify a unique, endocardially-derived angiogenic precursor cell population for coronary artery formation in mice and show that a DLL4/NOTCH1/VEGFA/VEGFR2 signaling axis is key for coronary artery development.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Fucosiltransferasas/genética , Neovascularización Fisiológica/genética , Transducción de Señal/genética , Animales , Proliferación Celular/genética , Enfermedad de la Arteria Coronaria/fisiopatología , Ecocardiografía , Fucosiltransferasas/deficiencia , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor 2 de Factores de Crecimiento Endotelial Vascular/deficiencia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/deficiencia , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
18.
J Biol Chem ; 292(38): 15964-15973, 2017 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-28729422

RESUMEN

Glycosylation in the endoplasmic reticulum (ER) is closely associated with protein folding and quality control. We recently described a non-canonical ER quality control mechanism for folding of thrombospondin type 1 repeats by protein O-fucosyltransferase 2 (POFUT2). Epidermal growth factor-like (EGF) repeats are also small cysteine-rich protein motifs that can be O-glycosylated by several ER-localized enzymes, including protein O-glucosyltransferase 1 (POGLUT1) and POFUT1. Both POGLUT1 and POFUT1 modify the Notch receptor on multiple EGF repeats and are essential for full Notch function. The fact that POGLUT1 and POFUT1 can distinguish between folded and unfolded EGF repeats raised the possibility that they participate in a quality control pathway for folding of EGF repeats in proteins such as Notch. Here, we demonstrate that cell-surface expression of endogenous Notch1 in HEK293T cells is dependent on the presence of POGLUT1 and POFUT1 in an additive manner. In vitro unfolding assays reveal that addition of O-glucose or O-fucose stabilizes a single EGF repeat and that addition of both O-glucose and O-fucose enhances stability in an additive manner. Finally, we solved the crystal structure of a single EGF repeat covalently modified by a full O-glucose trisaccharide at 2.2 Å resolution. The structure reveals that the glycan fills up a surface groove of the EGF with multiple contacts with the protein, providing a chemical basis for the stabilizing effects of the glycans. Taken together, this work suggests that O-fucose and O-glucose glycans cooperatively stabilize individual EGF repeats through intramolecular interactions, thereby regulating Notch trafficking in cells.


Asunto(s)
Factor de Crecimiento Epidérmico/química , Oxígeno/metabolismo , Receptores Notch/química , Receptores Notch/metabolismo , Secuencias Repetitivas de Aminoácido , Secuencia de Aminoácidos , Animales , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Glucosa/metabolismo , Glucosiltransferasas/deficiencia , Glucosiltransferasas/genética , Glicosilación , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Conformación Proteica , Transporte de Proteínas , Receptor Notch1/química , Receptor Notch1/metabolismo
19.
Int J Biochem Cell Biol ; 88: 44-54, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28483669

RESUMEN

Pulmonary fibrosis is a common outcome of a variety of pulmonary interstitial diseases, and myofibroblasts are the main culprit for this process. Recent studies have found that pericytes are one of the major sources of myofibroblasts; the transformation of which involves a complex process of activation of TGF-ß/Smad2/3 and PDGFß/Erk signaling pathways. We have reported that the transforming growth factor-ß receptor and platelet-derived growth factor-ß receptor (TGF-ßR I and PDGFßR, respectively) are modified by glycosylation. Thus, we hope to regulate the above-mentioned signal pathways through core fucosylation (CF) catalyzed by α-1,6-fucosyltransferase (FUT8). Previous work has confirmed that TGF-ß1 can induce the transformation of pericytes into myofibroblasts, while FUT8siRNA can inhibit such transformation. In the present study, we used an adenovirus packaging FUT8 shRNA to infect a bleomycin-induced pulmonary fibrosis mouse model and determined the effect of CF on pulmonary fibrosis by analyzing the mechanism of CF-mediated pericyte transformation. Our findings may shed new light on the mechanism of pulmonary interstitial fibrosis and provide a novel therapeutic target for clinical applications.


Asunto(s)
Fucosa/metabolismo , Pericitos/metabolismo , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Animales , Bleomicina/farmacología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glicosilación/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Pericitos/efectos de los fármacos , Pericitos/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Fibrosis Pulmonar/genética , ARN Interferente Pequeño/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
20.
Glycobiology ; 27(8): 777-786, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334865

RESUMEN

Protein O-fucosyltransferase-1 (POFUT1), which transfers fucose residues to acceptor sites on serine and threonine residues of epidermal growth factor-like repeats of recipient proteins, is essential for Notch signal transduction in mammals. Here, we examine the consequences of POFUT1 loss on the oncogenic signaling associated with certain leukemia-associated mutations of human Notch1, report the structures of human POFUT1 in free and GDP-fucose bound states, and assess the effects of Dowling-Degos mutations on human POFUT1 function. CRISPR-mediated knockout of POFUT1 in U2OS cells suppresses both normal Notch1 signaling, and the ligand-independent signaling associated with leukemogenic mutations of Notch1. Normal and oncogenic signaling are rescued by wild-type POFUT1 but rescue is impaired by an active-site R240A mutation. The overall structure of the human enzyme closely resembles that of the Caenorhabditis elegans protein, with an overall backbone RMSD of 0.93 Å, despite primary sequence identity of only 39% in the mature protein. GDP-fucose binding to the human enzyme induces limited backbone conformational movement, though the side chains of R43 and D244 reorient to make direct contact with the fucose moiety in the complex. The reported Dowling-Degos mutations of POFUT1, except for M262T, fail to rescue Notch1 signaling efficiently in the CRISPR-engineered POFUT1-/- background. Together, these studies identify POFUT1 as a potential target for cancers driven by Notch1 mutations and provide a structural roadmap for its inhibition.


Asunto(s)
Fucosiltransferasas/química , Fucosiltransferasas/genética , Hiperpigmentación/genética , Mutación , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Enfermedades Cutáneas Genéticas/genética , Enfermedades Cutáneas Papuloescamosas/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Fucosiltransferasas/deficiencia , Fucosiltransferasas/metabolismo , Humanos , Hiperpigmentación/metabolismo , Ligandos , Conformación Proteica , Enfermedades Cutáneas Genéticas/metabolismo , Enfermedades Cutáneas Papuloescamosas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA