Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(43): e2205277119, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36252012

RESUMEN

Mucins are the main macrocomponents of the mucus layer that protects the digestive tract from pathogens. Fucosylation of mucins increases mucus viscoelasticity and its resistance to shear stress. These properties are altered in patients with ulcerative colitis (UC), which is marked by a chronic inflammation of the distal part of the colon. Here, we show that levels of Fucosyltransferase 8 (FUT8) and specific mucins are increased in the distal inflamed colon of UC patients. Recapitulating this FUT8 overexpression in mucin-producing HT29-18N2 colonic cell line increases delivery of MUC1 to the plasma membrane and extracellular release of MUC2 and MUC5AC. Mucins secreted by FUT8 overexpressing cells are more resistant to removal from the cell surface than mucins secreted by FUT8-depleted cells (FUT8 KD). FUT8 KD causes intracellular accumulation of MUC1 and alters the ratio of secreted MUC2 to MUC5AC. These data fit well with the Fut8-/- mice phenotype, which are protected from UC. Fut8-/- mice exhibit a thinner proximal colon mucus layer with an altered ratio of neutral to acidic mucins. Together, our data reveal that FUT8 modifies the biophysical properties of mucus by controlling levels of cell surface MUC1 and quantity and quality of secreted MUC2 and MUC5AC. We suggest that these changes in mucus viscoelasticity likely facilitate bacterial-epithelial interactions leading to inflammation and UC progression.


Asunto(s)
Colitis Ulcerosa , Fucosiltransferasas , Animales , Ratones , Colitis Ulcerosa/genética , Colitis Ulcerosa/metabolismo , Fucosiltransferasas/genética , Inflamación , Mucina 2/genética , Mucina 2/metabolismo , Células HT29
2.
J Biol Chem ; 299(8): 105052, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37454739

RESUMEN

Chronic obstructive pulmonary disease (COPD), which includes emphysema and chronic bronchitis, is now the third cause of death worldwide, and COVID-19 infection has been reported as an exacerbation factor of them. In this study, we report that the intratracheal administration of the keratan sulfate-based disaccharide L4 mitigates the symptoms of elastase-induced emphysema in a mouse model. To know the molecular mechanisms, we performed a functional analysis of a C-type lectin receptor, langerin, a molecule that binds L4. Using mouse BMDCs (bone marrow-derived dendritic cells) as langerin-expressing cells, we observed the downregulation of IL-6 and TNFa and the upregulation of IL-10 after incubation with L4. We also identified CapG (a macrophage-capping protein) as a possible molecule that binds langerin by immunoprecipitation combined with a mass spectrometry analysis. We identified a portion of the CapG that was localized in the nucleus and binds to the promoter region of IL-6 and the TNFa gene in BMDCs, suggesting that CapG suppresses the gene expression of IL-6 and TNFa as an inhibitory transcriptional factor. To examine the effects of L4 in vivo, we also generated langerin-knockout mice by means of genome editing technology. In an emphysema mouse model, the administration of L4 did not mitigate the symptoms of emphysema as well as the inflammatory state of the lung in the langerin-knockout mice. These data suggest that the anti-inflammatory effect of L4 through the langerin-CapG axis represents a potential therapeutic target for the treatment of emphysema and COPD.


Asunto(s)
Disacáridos , Enfermedad Pulmonar Obstructiva Crónica , Enfisema Pulmonar , Animales , Ratones , Disacáridos/farmacología , Modelos Animales de Enfermedad , Interleucina-6/genética , Sulfato de Queratano/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfisema Pulmonar/tratamiento farmacológico , Enfisema Pulmonar/genética , Enfisema Pulmonar/inducido químicamente , Lectinas Tipo C/metabolismo
3.
J Biol Chem ; 299(12): 105365, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37865317

RESUMEN

Glycan structure is often modulated in disease or predisease states, suggesting that such changes might serve as biomarkers. Here, we generated a monoclonal antibody (mAb) against the core fucose of the N-glycan in human IgG. Notably, this mAb can be used in Western blotting and ELISA. ELISA using this mAb revealed a low level of the core fucose of the N-glycan in IgG, suggesting that the level of acore fucosylated (noncore fucosylated) IgG was increased in the sera of the patients with lung cancer, chronic obstructive pulmonary disease, and interstitial pneumonia compared to healthy subjects. In a coculture analysis using human lung adenocarcinoma A549 cells and antibody-secreting B cells, the downregulation of the FUT8 (α1,6 fucosyltransferase) gene and a low level of core fucose of the N-glycan in IgG in antibody-secreting B cells were observed after coculture. A dramatic alteration in gene expression profiles for cytokines, chemokines, and their receptors were also observed after coculturing, and we found that the identified C-C motif chemokine 2 was partially involved in the downregulation of the FUT8 gene and the low level of core fucose of the N-glycan in IgG in antibody-secreting B cells. We also developed a latex turbidimetric immunoassay using this mAb. These results suggest that communication with C-C motif chemokine 2 between lung cells and antibody-secreting B cells downregulate the level of core fucose of the N-glycan in IgG, i.e., the increased level of acore fucosylated (noncore fucosylated) IgG, which would be a novel biomarker for the diagnosis of patients with pulmonary diseases.


Asunto(s)
Anticuerpos Monoclonales , Fucosa , Inmunoglobulina G , Enfermedades Pulmonares , Polisacáridos , Humanos , Células A549 , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos , Linfocitos B/inmunología , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Fucosa/sangre , Fucosa/metabolismo , Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Técnicas de Inactivación de Genes , Inmunoensayo/normas , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Enfermedades Pulmonares/diagnóstico , Enfermedades Pulmonares/inmunología , Polisacáridos/metabolismo , Animales , Ratones , Células CHO , Células HEK293 , Cricetulus
4.
J Biol Chem ; 298(6): 101950, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35447118

RESUMEN

Asparagine-linked glycosylation (N-glycosylation) of proteins in the cancer secretome has been gaining increasing attention as a potential biomarker for cancer detection and diagnosis. Small extracellular vesicles (sEVs) constitute a large part of the cancer secretome, yet little is known about whether their N-glycosylation status reflects known cancer characteristics. Here, we investigated the N-glycosylation of sEVs released from small-cell lung carcinoma (SCLC) and non-small-cell lung carcinoma (NSCLC) cells. We found that the N-glycans of SCLC-sEVs were characterized by the presence of structural units also found in the brain N-glycome, while NSCLC-sEVs were dominated by typical lung-type N-glycans with NSCLC-associated core fucosylation. In addition, lectin-assisted N-glycoproteomics of SCLC-sEVs and NSCLC-sEVs revealed that integrin αV was commonly expressed in sEVs of both cancer cell types, while the epithelium-specific integrin α6ß4 heterodimer was selectively expressed in NSCLC-sEVs. Importantly, N-glycomics of the immunopurified integrin α6 from NSCLC-sEVs identified NSCLC-type N-glycans on this integrin subunit. Thus, we conclude that protein N-glycosylation in lung cancer sEVs may potentially reflect the histology of lung cancers.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Glicosilación , Neoplasias Pulmonares , Procesamiento Proteico-Postraduccional , Carcinoma Pulmonar de Células Pequeñas , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Vesículas Extracelulares/metabolismo , Humanos , Neoplasias Pulmonares/patología , Polisacáridos/metabolismo , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología
5.
J Biol Chem ; 296: 100354, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33524390

RESUMEN

Glycosylation, the most common posttranslational modification of proteins, is a stepwise process that relies on tight regulation of subcellular glycosyltransferase location to control the addition of each monosaccharide. Glycosyltransferases primarily reside and function in the endoplasmic reticulum (ER) and the Golgi apparatus; whether and how they traffic beyond the Golgi, how this trafficking is controlled, and how it impacts glycosylation remain unclear. Our previous work identified a connection between N-glycosylation and Rab11, a key player in the post-Golgi transport that connects recycling endosomes and other compartments. To learn more about the specific role of Rab11, we knocked down Rab11 in HeLa cells. Our findings indicate that Rab11 knockdown results in a dramatic enhancement in the sialylation of N-glycans. Structural analyses of glycans using lectins and LC-MS revealed that α2,3-sialylation is selectively enhanced, suggesting that an α2,3-sialyltransferase that catalyzes the sialyation of glycoproteins is activated or upregulated as the result of Rab11 knockdown. ST3GAL4 is the major α2,3-sialyltransferase that acts on N-glycans; we demonstrated that the localization of ST3GAL4, but not the levels of its mRNA, protein, or donor substrate, was altered by Rab11 depletion. In knockdown cells, ST3GAL4 is densely distributed in the trans-Golgi network, compared with the wider distribution in the Golgi and in other peripheral puncta in control cells, whereas the α2,6-sialyltransferase ST6GAL1 is predominantly localized to the Golgi regardless of Rab11 knockdown. This indicates that Rab11 may negatively regulate α2,3-sialylation by transporting ST3GAL4 to post-Golgi compartments (PGCs), which is a novel mechanism of glycosyltransferase regulation.


Asunto(s)
Sialiltransferasas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Glicosilación , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Transporte de Proteínas , Ratas , Red trans-Golgi/metabolismo
6.
Biochem Biophys Res Commun ; 633: 68-71, 2022 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-36344167

RESUMEN

Over the past 3 decades, our group has been involved in studies related to the biosynthesis of N-glycan branching and related glycosyltransferases and have purified most of these Golgi-derived enzymes to homogeneity using classical purification methods and cloned the cDNA of GnT-III, IV, V, VI and Fut8 except GnT-IX(Vb) which was obtained by homology cloning. Based primarily on our data, we briefly summarize the significance of three major enzymes and discuss perspectives for future studies on the occasion of Ernesto's 90th birthday celebration.


Asunto(s)
Enzima Ramificadora de 1,4-alfa-Glucano , Enfermedad de Alzheimer , Neoplasias , Enfermedad Pulmonar Obstructiva Crónica , Humanos , N-Acetilglucosaminiltransferasas/genética , Polisacáridos
7.
Biochem Soc Trans ; 49(1): 441-453, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33616615

RESUMEN

Glycosylation represents one of the most abundant posttranslational modification of proteins. Glycosylation products are diverse and are regulated by the cooperative action of various glycosyltransferases, glycosidases, substrates thereof: nucleoside sugars and their transporters, and chaperons. In this article, we focus on a glycosyltransferase, α1,6-fucosyltransferase (Fut8) and its product, the core fucose structure on N-glycans, and summarize the potential protective functions of this structure against emphysema and chronic obstructive pulmonary disease (COPD). Studies of FUT8 and its enzymatic product, core fucose, are becoming an emerging area of interest in various fields of research including inflammation, cancer and therapeutics. This article discusses what we can learn from studies of Fut8 and core fucose by using knockout mice or in vitro studies that were conducted by our group as well as other groups. We also include a discussion of the potential protective functions of the keratan sulfate (KS) disaccharide, namely L4, against emphysema and COPD as a glycomimetic. Glycomimetics using glycan analogs is one of the more promising therapeutics that compensate for the usual therapeutic strategy that involves targeting the genome and the proteome. These typical glycans using KS derivatives as glycomimetics, will likely become a clue to the development of novel and effective therapeutic strategies.


Asunto(s)
Materiales Biomiméticos/uso terapéutico , Sulfato de Queratano/química , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Antígenos de Superficie/fisiología , Materiales Biomiméticos/química , Fucosa/metabolismo , Fucosiltransferasas/fisiología , Glicosilación , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Lectinas Tipo C/fisiología , Lectinas de Unión a Manosa/antagonistas & inhibidores , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/metabolismo , Lectinas de Unión a Manosa/fisiología , Ratones , Ratones Noqueados , Terapia Molecular Dirigida/métodos , Polisacáridos/química , Polisacáridos/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo
8.
Adv Exp Med Biol ; 1325: 137-149, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34495533

RESUMEN

Extracellular vesicles (EVs), a generic term for any vesicles or particles that are released from cells, play an important role in modulating numerous biological and pathological events, including development, differentiation, aging, thrombus formation, immune responses, neurodegenerative diseases, and tumor progression. During the biogenesis of EVs, they encapsulate biologically active macromolecules (i.e., nucleotides and proteins) and transmit signals for delivering them to neighboring or cells that are located some distance away. In contrast, there are receptor molecules on the surface of EVs that function to mediate EV-to-cell and EV-to-matrix interactions. A growing body of evidence indicates that the EV surface is heavily modified with glycans, the function of which is to regulate the biogenesis and extracellular behaviors of EVs. In this chapter, we introduce the current status of our knowledge concerning EV glycosylation and discuss how it influences EV biology, highlighting the potential roles of EV glycans in clinical applications.


Asunto(s)
Exosomas , Vesículas Extracelulares , Enfermedades Neurodegenerativas , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Glicosilación , Humanos , Enfermedades Neurodegenerativas/metabolismo
9.
Int J Mol Sci ; 22(16)2021 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-34445285

RESUMEN

N-glycosylation is essential for many biological processes in mammals. A variety of N-glycan structures exist, of which, the formation of bisecting N-acetylglucosamine (GlcNAc) is catalyzed by N-acetylglucosaminyltransferase-III (GnT-III, encoded by the Mgat3 gene). We previously identified various bisecting GlcNAc-modified proteins involved in Alzheimer's disease and cancer. However, the mechanisms by which GnT-III acts on the target proteins are unknown. Here, we performed comparative glycoproteomic analyses using brain membranes of wild type (WT) and Mgat3-deficient mice. Target glycoproteins of GnT-III were enriched with E4-phytohemagglutinin (PHA) lectin, which recognizes bisecting GlcNAc, and analyzed by liquid chromatograph-mass spectrometry. We identified 32 N-glycosylation sites (Asn-Xaa-Ser/Thr, Xaa ≠ Pro) that were modified with bisecting GlcNAc. Sequence alignment of identified N-glycosylation sites that displayed bisecting GlcNAc suggested that GnT-III does not recognize a specific primary amino acid sequence. The molecular modeling of GluA1 as one of the good cell surface substrates for GnT-III in the brain, indicated that GnT-III acts on N-glycosylation sites located in a highly flexible and mobile loop of GluA1. These results suggest that the action of GnT-III is partially affected by the tertiary structure of target proteins, which can accommodate bisecting GlcNAc that generates a bulky flipped-back conformation of the modified glycans.


Asunto(s)
Acetilglucosamina/metabolismo , Encéfalo/metabolismo , Membrana Celular/metabolismo , Péptidos/metabolismo , Receptores AMPA/metabolismo , Análisis de Secuencia de Proteína , Acetilglucosamina/genética , Animales , Membrana Celular/genética , Glicosilación , Ratones , Ratones Noqueados , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/metabolismo , Mapeo Peptídico , Péptidos/genética , Receptores AMPA/genética
10.
PLoS Genet ; 13(4): e1006696, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28426790

RESUMEN

The cytoplasmic peptide:N-glycanase (Ngly1 in mammals) is a de-N-glycosylating enzyme that is highly conserved among eukaryotes. It was recently reported that subjects harboring mutations in the NGLY1 gene exhibited severe systemic symptoms (NGLY1-deficiency). While the enzyme obviously has a critical role in mammals, its precise function remains unclear. In this study, we analyzed Ngly1-deficient mice and found that they are embryonic lethal in C57BL/6 background. Surprisingly, the additional deletion of the gene encoding endo-ß-N-acetylglucosaminidase (Engase), which is another de-N-glycosylating enzyme but leaves a single GlcNAc at glycosylated Asn residues, resulted in the partial rescue of the lethality of the Ngly1-deficient mice. Additionally, we also found that a change in the genetic background of C57BL/6 mice, produced by crossing the mice with an outbred mouse strain (ICR) could partially rescue the embryonic lethality of Ngly1-deficient mice. Viable Ngly1-deficient mice in a C57BL/6 and ICR mixed background, however, showed a very severe phenotype reminiscent of the symptoms of NGLY1-deficiency subjects. Again, many of those defects were strongly suppressed by the additional deletion of Engase in the C57BL/6 and ICR mixed background. The defects observed in Ngly1/Engase-deficient mice (C57BL/6 background) and Ngly1-deficient mice (C57BL/6 and ICR mixed background) closely resembled some of the symptoms of patients with an NGLY1-deficiency. These observations strongly suggest that the Ngly1- or Ngly1/Engase-deficient mice could serve as a valuable animal model for studies related to the pathogenesis of the NGLY1-deficiency, and that cytoplasmic ENGase represents one of the potential therapeutic targets for this genetic disorder.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Animales , Citoplasma/enzimología , Enfermedades Genéticas Congénitas/terapia , Glicosilación , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Eliminación de Secuencia/genética
11.
Anal Biochem ; 580: 21-29, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31173727

RESUMEN

Tumor cells secrete membrane vesicles of various sizes, termed extracellular vesicles (EVs), which have gained increasing attention as potential tumor diagnostic markers. Tumor-derived EVs are enriched with high-mannose-type glycans. Here, we report the affinity isolation of EVs from human melanoma A375 cells by using high-mannose-type glycan-specific agglutinin from Oscillatoria Agardhii (OAA). Glycan analysis of melanoma EVs revealed the presence of high-mannose-type glycans with structural units preferred by OAA. We showed that in solution, OAA binds to melanoma EVs in a high-mannose-type glycan-dependent manner. Furthermore, OAA-immobilized beads were found to capture 60% of the particles and most proteinous components from melanoma EVs. Major EV glycoproteins that potentially interact with OAA were identified to be cluster of differentiation 109 (CD109), integrin α6 and a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10). In addition to melanoma EVs, OAA captured EVs from human lung cancer, glioblastoma and colon cancer cells, but not those from endothelial cells and fibroblasts. These results indicate that OAA-immobilized beads may serve as a novel platform for affinity-capture of tumor-derived EVs.


Asunto(s)
Vesículas Extracelulares/metabolismo , Lectinas de Unión a Manosa/metabolismo , Polisacáridos/metabolismo , Células A549 , Proteínas Bacterianas/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Células HCT116 , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo , Oscillatoria/metabolismo , Unión Proteica
12.
Int J Mol Sci ; 20(23)2019 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-31810196

RESUMEN

Oligosaccharyltransferase (OST) is a multi-span membrane protein complex that catalyzes the addition of glycans to selected Asn residues within nascent polypeptides in the lumen of the endoplasmic reticulum. This process, termed N-glycosylation, is a fundamental post-translational protein modification that is involved in the quality control, trafficking of proteins, signal transduction, and cell-to-cell communication. Given these crucial roles, N-glycosylation is essential for homeostasis at the systemic and cellular levels, and a deficiency in genes that encode for OST subunits often results in the development of complex genetic disorders. A growing body of evidence has also demonstrated that the expression of OST subunits is cell context-dependent and is frequently altered in malignant cells, thus contributing to tumor cell survival and proliferation. Importantly, a recently developed inhibitor of OST has revealed this enzyme as a potential target for the treatment of incurable drug-resistant tumors. This review summarizes our current knowledge regarding the functions of OST in the light of health and tumor progression, and discusses perspectives on the clinical relevance of inhibiting OST as a tumor treatment.


Asunto(s)
Resistencia a Antineoplásicos/genética , Hexosiltransferasas/genética , Proteínas de la Membrana/genética , Neoplasias/genética , Procesamiento Proteico-Postraduccional/genética , Secuencia de Aminoácidos/genética , Asparagina/genética , Progresión de la Enfermedad , Retículo Endoplásmico/genética , Glicosilación , Hexosiltransferasas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Neoplasias/tratamiento farmacológico , Polisacáridos/genética
13.
Proc Natl Acad Sci U S A ; 112(5): 1398-403, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25605922

RESUMEN

The cytoplasmic peptide:N-glycanase (PNGase; Ngly1 in mice) is a deglycosylating enzyme involved in the endoplasmic reticulum (ER)-associated degradation (ERAD) process. The precise role of Ngly1 in the ERAD process, however, remains unclear in mammals. The findings reported herein, using mouse embryonic fibroblast (MEF) cells, that the ablation of Ngly1 causes dysregulation of the ERAD process. Interestingly, not only delayed degradation but also the deglycosylation of a misfolded glycoprotein was observed in Ngly1(-/-) MEF cells. The unconventional deglycosylation reaction was found to be catalyzed by the cytosolic endo-ß-N-acetylglucosaminidase (ENGase), generating aggregation-prone N-GlcNAc proteins. The ERAD dysregulation in cells lacking Ngly1 was restored by the additional knockout of ENGase gene. Thus, our study underscores the functional importance of Ngly1 in the ERAD process and provides a potential mechanism underlying the phenotypic consequences of a newly emerging genetic disorder caused by mutation of the human NGLY1 gene.


Asunto(s)
Acetilglucosamina/metabolismo , Retículo Endoplásmico/metabolismo , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/metabolismo , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Animales , Células Cultivadas , Ratones , Mutación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Ensayo de Radioinmunoprecipitación
14.
J Biol Chem ; 291(15): 8048-58, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-26858256

RESUMEN

Phosphorylated oligosaccharides (POSs) are produced by the degradation of dolichol-linked oligosaccharides (DLOs) by an unclarified mechanism in mammalian cells. Although POSs are exclusively found in the cytosol, their intracellular fates remain unclear. Our findings indicate that POSs are catabolized via a non-lysosomal glycan degradation pathway that involves a cytosolic endo-ß-N-acetylglucosaminidase (ENGase). Quantitative and structural analyses of POSs revealed that ablation of the ENGase results in the significant accumulation of POSs with a hexasaccharide structure composed of Manα1,2Manα1,3(Manα1,6)Manß1,4GlcNAcß1,4GlcNAc.In vitroENGase assays revealed that the presence of an α1,2-linked mannose residue facilitates the hydrolysis of POSs by the ENGase. Liquid chromatography-mass spectrometric analyses and fluorescent labeling experiments show that such POSs contain one phosphate group at the reducing end. These results indicate that ENGase efficiently hydrolyzes POSs that are larger than Man4GlcNAc2-P, generating GlcNAc-1-P and neutral Gn1-type free oligosaccharides. These results provide insight into important aspects of the generation and degradation of POSs.


Asunto(s)
Citosol/metabolismo , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/metabolismo , Oligosacáridos/metabolismo , Animales , Secuencia de Carbohidratos , Línea Celular , Hidrólisis , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Oligosacáridos/química , Fosforilación
15.
Cell Mol Life Sci ; 72(13): 2509-33, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25772500

RESUMEN

Asparagine (N)-linked protein glycosylation, which takes place in the eukaryotic endoplasmic reticulum (ER), is important for protein folding, quality control and the intracellular trafficking of secretory and membrane proteins. It is known that, during N-glycosylation, considerable amounts of lipid-linked oligosaccharides (LLOs), the glycan donor substrates for N-glycosylation, are hydrolyzed to form free N-glycans (FNGs) by unidentified mechanisms. FNGs are also generated in the cytosol by the enzymatic deglycosylation of misfolded glycoproteins during ER-associated degradation. FNGs derived from LLOs and misfolded glycoproteins are eventually merged into one pool in the cytosol and the various glycan structures are processed to a near homogenous glycoform. This article summarizes the current state of our knowledge concerning the formation and catabolism of FNGs.


Asunto(s)
Asparagina/metabolismo , Vías Biosintéticas/fisiología , Retículo Endoplásmico/metabolismo , Modelos Biológicos , Polisacáridos/biosíntesis , Polisacáridos/metabolismo , Glicosilación , Hidrólisis , Lipopolisacáridos/metabolismo , Pliegue de Proteína
16.
Biosci Biotechnol Biochem ; 80(1): 152-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26264652

RESUMEN

In the cytosol of Saccharomyces cerevisiae, most of the free N-glycans (FNGs) are generated from misfolded glycoproteins by the action of the cytoplasmic peptide: N-glycanase (Png1). A cytosol/vacuole α-mannosidase, Ams1, then trims the FNGs to eventually form a trisaccharide composed of Manß1,4GlcNAc ß1,4GlcNAc (Man1GlcNAc2). Whether or not the resulting Man1GlcNAc2 is enzymatically degraded further, however, is currently unknown. The objective of this study was to unveil the fate of Man1GlcNAc2 in S. cerevisiae. Quantitative analyses of the FNGs revealed a steady increase in the amount of Man1GlcNAc2 produced in the post-diauxic and stationary phases, suggesting that this trisaccharide is not catabolized during this period. Inoculation of the stationary phase cells into fresh medium resulted in a reduction in the levels of Man1GlcNAc2. However, this reduction was caused by its dilution due to cell division in the fresh medium. Our results thus indicate that Man1GlcNAc2 is not enzymatically catabolized in S. cerevisiae.


Asunto(s)
Regulación Fúngica de la Expresión Génica , Saccharomyces cerevisiae/genética , Trisacáridos/metabolismo , Secuencia de Carbohidratos , Citosol/metabolismo , Glicosilación , Metabolismo , Datos de Secuencia Molecular , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Polisacáridos/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , alfa-Manosidasa/genética , alfa-Manosidasa/metabolismo
17.
Proc Natl Acad Sci U S A ; 110(48): 19366-71, 2013 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-24218558

RESUMEN

The glycolipid Glc3Man9GlcNAc2-pyrophosphate-dolichol serves as the precursor for asparagine (N)-linked protein glycosylation in mammals. The biosynthesis of dolichol-linked oligosaccharides (DLOs) is arrested in low-glucose environments via unknown mechanisms, resulting in abnormal N-glycosylation. Here, we show that under glucose deprivation, DLOs are prematurely degraded during the early stages of DLO biosynthesis by pyrophosphatase, leading to the release of singly phosphorylated oligosaccharides into the cytosol. We identified that the level of GDP-mannose (Man), which serves as a donor substrate for DLO biosynthesis, is substantially reduced under glucose deprivation. We provide evidence that the selective shutdown of the GDP-Man biosynthetic pathway is sufficient to induce the release of phosphorylated oligosaccharides. These results indicate that glucose-regulated metabolic changes in the GDP-Man biosynthetic pathway cause the biosynthetic arrest of DLOs and facilitate their premature degradation by pyrophosphatase. We propose that this degradation system may avoid abnormal N-glycosylation with premature oligosaccharides under conditions that impair efficient DLO biosynthesis.


Asunto(s)
Asparagina/metabolismo , Vías Biosintéticas/fisiología , Dolicoles/metabolismo , Retículo Endoplásmico/metabolismo , Guanosina Difosfato Manosa/biosíntesis , Oligosacáridos/biosíntesis , Péptidos/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Citosol/metabolismo , Fibroblastos , Glucosa/deficiencia , Glicosilación , Ratones , Modelos Biológicos , Oligosacáridos/metabolismo , Polisacáridos/metabolismo , Pirofosfatasas/metabolismo , Espectrometría de Masas en Tándem
18.
Glycobiology ; 25(11): 1196-205, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26206502

RESUMEN

During asparagine (N)-linked protein glycosylation, eukaryotic cells generate considerable amounts of free oligosaccharides (fOSs) in the cytosol. It is generally assumed that such fOSs are produced by the deglycosylation of misfolded N-glycoproteins that are destined for proteasomal degradation or as the result of the degradation of dolichol-linked oligosaccharides (DLOs), which serve as glycan donor substrates in N-glycosylation reactions. The findings reported herein show that the majority of cytosolic fOSs are generated by a peptide:N-glycanase (PNGase) and an endo-ß-N-acetylglucosaminidase (ENGase)-independent pathway in mammalian cells. The ablation of the cytosolic deglycosylating enzymes, PNGase and ENGase, in mouse embryonic fibroblasts had little effect on the amount of cytosolic fOSs generated. Quantitative analyses of fOSs using digitonin-permeabilized cells revealed that they are generated by the degradation of fully assembled Glc3Man9GlcNAc2-pyrophosphate-dolichol (PP-Dol) in the lumen of the endoplasmic reticulum. Because the degradation of Glc3Man9GlcNAc2-PP-Dol is greatly inhibited in the presence of an N-glycosylation acceptor peptide that is recognized by the oligosaccharyltransferase (OST), the OST-mediated hydrolysis of DLO is the most likely mechanism responsible for the production of a large fraction of the cytosolic fOSs.


Asunto(s)
Citosol/metabolismo , Dolicoles/metabolismo , Oligosacáridos/metabolismo , Acetilglucosaminidasa/metabolismo , Animales , Células Cultivadas , Retículo Endoplásmico/metabolismo , Fibroblastos/metabolismo , Glicoproteínas/metabolismo , Glicósido Hidrolasas/metabolismo , Hexosiltransferasas/metabolismo , Hidrólisis , Proteínas de la Membrana/metabolismo , Ratones
19.
J Biol Chem ; 288(45): 32673-32684, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24062310

RESUMEN

Asparagine (N)-linked glycosylation regulates numerous cellular activities, such as glycoprotein quality control, intracellular trafficking, and cell-cell communications. In eukaryotes, the glycosylation reaction is catalyzed by oligosaccharyltransferase (OST), a multimembrane protein complex that is localized in the endoplasmic reticulum (ER). During N-glycosylation in the ER, the protein-unbound form of oligosaccharides (free oligosaccharides; fOSs), which is structurally related to N-glycan, is released into the ER lumen. However, the enzyme responsible for this process remains unidentified. Here, we demonstrate that eukaryotic OST generates fOSs. Biochemical and genetic analyses using mutant strains of Saccharomyces cerevisiae revealed that the generation of fOSs is tightly correlated with the N-glycosylation activity of OST. Furthermore, we present evidence that the purified OST complex can generate fOSs by hydrolyzing dolichol-linked oligosaccharide, the glycan donor substrate for N-glycosylation. The heterologous expression of a single subunit of OST from the protozoan Leishmania major in S. cerevisiae demonstrated that this enzyme functions both in N-glycosylation and generation of fOSs. This study provides insight into the mechanism of PNGase-independent formation of fOSs.


Asunto(s)
Hexosiltransferasas/metabolismo , Leishmania major/enzimología , Proteínas de la Membrana/metabolismo , Oligosacáridos/metabolismo , Proteínas Protozoarias/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Glicosilación , Hexosiltransferasas/genética , Leishmania major/genética , Proteínas de la Membrana/genética , Oligosacáridos/genética , Oligosacáridos de Poliisoprenil Fosfato/metabolismo , Proteínas Protozoarias/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
20.
J Biol Chem ; 288(37): 26898-907, 2013 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-23880766

RESUMEN

Macroautophagy is an essential, homeostatic process involving degradation of a cell's own components; it plays a role in catabolizing cellular components, such as protein or lipids, and damaged or excess organelles. Here, we show that in Atg5(-/-) cells, sialyloligosaccharides specifically accumulated in the cytosol. Accumulation of these glycans was observed under non-starved conditions, suggesting that non-induced, basal autophagy is essential for their catabolism. Interestingly, once accumulated in the cytosol, sialylglycans cannot be efficiently catabolized by resumption of the autophagic process, suggesting that functional autophagy is important for preventing sialyloligosaccharides from accumulating in the cytosol. Moreover, knockdown of sialin, a lysosomal transporter of sialic acids, resulted in a significant reduction of sialyloligosaccharides, implying that autophagy affects the substrate specificity of this transporter. This study thus provides a surprising link between basal autophagy and catabolism of N-linked glycans.


Asunto(s)
Autofagia , Regulación de la Expresión Génica , Proteínas Asociadas a Microtúbulos/genética , Oligosacáridos/metabolismo , Animales , Proteína 5 Relacionada con la Autofagia , Citosol/metabolismo , Fibroblastos/citología , Lisosomas/metabolismo , Metabolismo , Ratones , Oligosacáridos/genética , ARN Interferente Pequeño/metabolismo , Fracciones Subcelulares/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA