Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(47): 29595-29601, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33154157

RESUMEN

Mammalian protein N-linked glycosylation is critical for glycoprotein folding, quality control, trafficking, recognition, and function. N-linked glycans are synthesized from Glc3Man9GlcNAc2 precursors that are trimmed and modified in the endoplasmic reticulum (ER) and Golgi apparatus by glycoside hydrolases and glycosyltransferases. Endo-α-1,2-mannosidase (MANEA) is the sole endo-acting glycoside hydrolase involved in N-glycan trimming and is located within the Golgi, where it allows ER-escaped glycoproteins to bypass the classical N-glycosylation trimming pathway involving ER glucosidases I and II. There is considerable interest in the use of small molecules that disrupt N-linked glycosylation as therapeutic agents for diseases such as cancer and viral infection. Here we report the structure of the catalytic domain of human MANEA and complexes with substrate-derived inhibitors, which provide insight into dynamic loop movements that occur on substrate binding. We reveal structural features of the human enzyme that explain its substrate preference and the mechanistic basis for catalysis. These structures have inspired the development of new inhibitors that disrupt host protein N-glycan processing of viral glycans and reduce the infectivity of bovine viral diarrhea and dengue viruses in cellular models. These results may contribute to efforts aimed at developing broad-spectrum antiviral agents and help provide a more in-depth understanding of the biology of mammalian glycosylation.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Glicosilación/efectos de los fármacos , Manosidasas/química , Manosidasas/farmacología , Animales , Diarrea Mucosa Bovina Viral/tratamiento farmacológico , Bovinos , Línea Celular , Virus del Dengue/efectos de los fármacos , Perros , Glucosidasas/metabolismo , Humanos , Células de Riñón Canino Madin Darby , Polisacáridos/metabolismo , Vías Secretoras/efectos de los fármacos
2.
Chemistry ; 27(44): 11291-11297, 2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34106504

RESUMEN

Mucopolysaccharidosis type IIIB is a devastating neurological disease caused by a lack of the lysosomal enzyme, α-N-acetylglucosaminidase (NAGLU), leading to a toxic accumulation of heparan sulfate. Herein we explored a pharmacological chaperone approach to enhance the residual activity of NAGLU in patient fibroblasts. Capitalizing on the three-dimensional structures of two modest homoiminosugar-based NAGLU inhibitors in complex with bacterial homolog of NAGLU, CpGH89, we have synthesized a library of 17 iminosugar C-glycosides mimicking N-acetyl-D-glucosamine and bearing various pseudo-anomeric substituents of both α- and ß-configuration. Elaboration of the aglycon moiety results in low micromolar selective inhibitors of human recombinant NAGLU, but surprisingly it is the non-functionalized and wrongly configured ß-homoiminosugar that was proved to act as the most promising pharmacological chaperone, promoting a 2.4 fold activity enhancement of mutant NAGLU at its optimal concentration.


Asunto(s)
Mucopolisacaridosis III , Acetilglucosaminidasa , Glicósidos , Humanos , Enfermedades Raras
3.
Proc Natl Acad Sci U S A ; 114(32): 8544-8549, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28739903

RESUMEN

Glycoproteins traversing the eukaryotic secretory pathway begin life in the endoplasmic reticulum (ER), where their folding is surveyed by the 170-kDa UDP-glucose:glycoprotein glucosyltransferase (UGGT). The enzyme acts as the single glycoprotein folding quality control checkpoint: it selectively reglucosylates misfolded glycoproteins, promotes their association with ER lectins and associated chaperones, and prevents premature secretion from the ER. UGGT has long resisted structural determination and sequence-based domain boundary prediction. Questions remain on how this single enzyme can flag misfolded glycoproteins of different sizes and shapes for ER retention and how it can span variable distances between the site of misfold and a glucose-accepting N-linked glycan on the same glycoprotein. Here, crystal structures of a full-length eukaryotic UGGT reveal four thioredoxin-like (TRXL) domains arranged in a long arc that terminates in two ß-sandwiches tightly clasping the glucosyltransferase domain. The fold of the molecule is topologically complex, with the first ß-sandwich and the fourth TRXL domain being encoded by nonconsecutive stretches of sequence. In addition to the crystal structures, a 15-Å cryo-EM reconstruction reveals interdomain flexibility of the TRXL domains. Double cysteine point mutants that engineer extra interdomain disulfide bridges rigidify the UGGT structure and exhibit impaired activity. The intrinsic flexibility of the TRXL domains of UGGT may therefore endow the enzyme with the promiscuity needed to recognize and reglucosylate its many different substrates and/or enable reglucosylation of N-linked glycans situated at variable distances from the site of misfold.


Asunto(s)
Glucosiltransferasas/química , Glucosiltransferasas/fisiología , Animales , Chaetomium/genética , Chaetomium/metabolismo , Cristalografía por Rayos X/métodos , Retículo Endoplásmico/metabolismo , Eucariontes/metabolismo , Células Eucariotas/metabolismo , Glucosiltransferasas/metabolismo , Glicoproteínas/metabolismo , Conformación Molecular , Dominios Proteicos/fisiología , Pliegue de Proteína , Transporte de Proteínas/fisiología , Especificidad por Sustrato
4.
Proc Natl Acad Sci U S A ; 113(32): E4630-8, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27462106

RESUMEN

The biosynthesis of enveloped viruses depends heavily on the host cell endoplasmic reticulum (ER) glycoprotein quality control (QC) machinery. This dependency exceeds the dependency of host glycoproteins, offering a window for the targeting of ERQC for the development of broad-spectrum antivirals. We determined small-angle X-ray scattering (SAXS) and crystal structures of the main ERQC enzyme, ER α-glucosidase II (α-GluII; from mouse), alone and in complex with key ligands of its catalytic cycle and antiviral iminosugars, including two that are in clinical trials for the treatment of dengue fever. The SAXS data capture the enzyme's quaternary structure and suggest a conformational rearrangement is needed for the simultaneous binding of a monoglucosylated glycan to both subunits. The X-ray structures with key catalytic cycle intermediates highlight that an insertion between the +1 and +2 subsites contributes to the enzyme's activity and substrate specificity, and reveal that the presence of d-mannose at the +1 subsite renders the acid catalyst less efficient during the cleavage of the monoglucosylated substrate. The complexes with iminosugar antivirals suggest that inhibitors targeting a conserved ring of aromatic residues between the α-GluII +1 and +2 subsites would have increased potency and selectivity, thus providing a template for further rational drug design.


Asunto(s)
Antivirales/farmacología , Retículo Endoplásmico/enzimología , Inhibidores de Glicósido Hidrolasas/farmacología , alfa-Glucosidasas/química , Animales , Catálisis , Cristalografía por Rayos X , Ratones , Conformación Proteica , Subunidades de Proteína , Dispersión del Ángulo Pequeño , Especificidad por Sustrato
5.
Adv Exp Med Biol ; 1062: 265-276, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29845539

RESUMEN

Targeting the host-cell endoplasmic reticulum quality control (ERQC) pathway is an effective broad-spectrum antiviral strategy. The two ER resident α-glucosidases whose sequential action permits entry in this pathway are the targets of glucomimetic inhibitors. Knowledge of the molecular details of the ER α-glucosidase II (α-Glu II) structure was limited. We determined crystal structures of a trypsinolytic fragment of murine α-Glu II, alone and in complex with key catalytic cycle ligands, and four different broad-spectrum antiviral iminosugar inhibitors, two of which are currently in clinical trials against dengue fever. The structures highlight novel portions of the enzyme outside its catalytic pocket which contribute to its activity and substrate specificity. These crystal structures and hydrogen-deuterium exchange mass spectrometry of the murine ER alpha glucosidase II heterodimer uncover the quaternary arrangement of the enzyme's α- and ß-subunits, and suggest a conformational rearrangement of ER α-Glu II upon association of the enzyme with client glycoproteins.


Asunto(s)
Retículo Endoplásmico/enzimología , Virosis/enzimología , Virosis/inmunología , Fenómenos Fisiológicos de los Virus , alfa-Glucosidasas/química , alfa-Glucosidasas/inmunología , Animales , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/virología , Interacciones Huésped-Patógeno , Humanos , Virosis/genética , Virosis/virología , Virus/genética , alfa-Glucosidasas/genética
6.
Biochem Soc Trans ; 45(2): 571-582, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28408497

RESUMEN

Many viruses require the host endoplasmic reticulum protein-folding machinery in order to correctly fold one or more of their glycoproteins. Iminosugars with glucose stereochemistry target the glucosidases which are key for entry into the glycoprotein folding cycle. Viral glycoproteins are thus prevented from interacting with the protein-folding machinery leading to misfolding and an antiviral effect against a wide range of different viral families. As iminosugars target host enzymes, they should be refractory to mutations in the virus. Iminosugars therefore have great potential for development as broad-spectrum antiviral therapeutics. We outline the mechanism giving rise to the antiviral activity of iminosugars, the current progress in the development of iminosugar antivirals and future prospects for this field.


Asunto(s)
Antivirales/farmacología , Glucosidasas/antagonistas & inhibidores , Iminoazúcares/farmacología , Animales , Antivirales/química , Antivirales/uso terapéutico , Ensayos Clínicos como Asunto , Enfermedades Transmisibles/tratamiento farmacológico , Enfermedades Transmisibles/virología , Retículo Endoplásmico/enzimología , Humanos , Iminoazúcares/química , Iminoazúcares/uso terapéutico , Pliegue de Proteína/efectos de los fármacos , Proteínas Virales/química
7.
Proc Natl Acad Sci U S A ; 109(3): 781-6, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22219371

RESUMEN

N-linked glycans play key roles in protein folding, stability, and function. Biosynthetic modification of N-linked glycans, within the endoplasmic reticulum, features sequential trimming and readornment steps. One unusual enzyme, endo-α-mannosidase, cleaves mannoside linkages internally within an N-linked glycan chain, short circuiting the classical N-glycan biosynthetic pathway. Here, using two bacterial orthologs, we present the first structural and mechanistic dissection of endo-α-mannosidase. Structures solved at resolutions 1.7-2.1 Å reveal a (ß/α)(8) barrel fold in which the catalytic center is present in a long substrate-binding groove, consistent with cleavage within the N-glycan chain. Enzymatic cleavage of authentic Glc(1/3)Man(9)GlcNAc(2) yields Glc(1/3)-Man. Using the bespoke substrate α-Glc-1,3-α-Man fluoride, the enzyme was shown to act with retention of anomeric configuration. Complexes with the established endo-α-mannosidase inhibitor α-Glc-1,3-deoxymannonojirimycin and a newly developed inhibitor, α-Glc-1,3-isofagomine, and with the reducing-end product α-1,2-mannobiose structurally define the -2 to +2 subsites of the enzyme. These structural and mechanistic data provide a foundation upon which to develop new enzyme inhibitors targeting the hijacking of N-glycan synthesis in viral disease and cancer.


Asunto(s)
Bacteroides/enzimología , Polisacáridos/química , Polisacáridos/metabolismo , alfa-Manosidasa/metabolismo , Biocatálisis , Conformación de Carbohidratos , Dominio Catalítico , Secuencia Conservada , Humanos , Cinética , Ligandos , Modelos Moleculares , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Electricidad Estática , alfa-Manosidasa/antagonistas & inhibidores
8.
J Inherit Metab Dis ; 37(2): 297-308, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24136589

RESUMEN

BACKGROUND: UDP-GlcNAc 2-epimerase/ManNAc 6-kinase (GNE) is a bifunctional enzyme responsible for the first committed steps in the synthesis of sialic acid, a common terminal monosaccharide in both protein and lipid glycosylation. GNE mutations are responsible for a rare autosomal recessive neuromuscular disorder, GNE myopathy (also called hereditary inclusion body myopathy). The connection between the impairment of sialic acid synthesis and muscle pathology in GNE myopathy remains poorly understood. METHODS: Glycosphingolipid (GSL) analysis was performed by HPLC in multiple models of GNE myopathy, including patients' fibroblasts and plasma, control fibroblasts with inhibited GNE epimerase activity through a novel imino sugar, and tissues of Gne(M712T/M712T) knock-in mice. RESULTS: Not only neutral GSLs, but also sialylated GSLs, were significantly increased compared to controls in all tested models of GNE myopathy. Treatment of GNE myopathy fibroblasts with N-acetylmannosamine (ManNAc), a sialic acid precursor downstream of GNE epimerase activity, ameliorated the increased total GSL concentrations. CONCLUSION: GNE myopathy models have increased total GSL concentrations. ManNAc supplementation results in decrease of GSL levels, linking abnormal increase of total GSLs in GNE myopathy to defects in the sialic acid biosynthetic pathway. These data advocate for further exploring GSL concentrations as an informative biomarker, not only for GNE myopathy, but also for other disorders of sialic acid metabolism.


Asunto(s)
Glicoesfingolípidos/metabolismo , Complejos Multienzimáticos/metabolismo , Enfermedades Musculares/metabolismo , Animales , Estudios de Casos y Controles , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Glicoesfingolípidos/sangre , Glicoesfingolípidos/genética , Hexosaminas/sangre , Hexosaminas/genética , Hexosaminas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Complejos Multienzimáticos/sangre , Complejos Multienzimáticos/genética , Músculos/metabolismo , Enfermedades Musculares/sangre , Enfermedades Musculares/genética , Mutación , Ácido N-Acetilneuramínico/sangre , Ácido N-Acetilneuramínico/genética , Ácido N-Acetilneuramínico/metabolismo
9.
Cell Mol Life Sci ; 70(15): 2799-814, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23503623

RESUMEN

Endoplasmic reticulum-associated degradation (ERAD) is a key cellular process whereby misfolded proteins are removed from the endoplasmic reticulum (ER) for subsequent degradation by the ubiquitin/proteasome system. In the present work, analysis of the released, free oligosaccharides (FOS) derived from all glycoproteins undergoing ERAD, has allowed a global estimation of the mechanisms of this pathway rather than following model proteins through degradative routes. Examining the FOS produced in endomannosidase-compromised cells following α-glucosidase inhibition has revealed a mechanism for clearing Golgi-retrieved glycoproteins that have failed to enter the ER quality control cycle. The Glc3Man7GlcNAc2 FOS species has been shown to be produced in the ER lumen by a mechanism involving a peptide: N-glycanase-like activity, and its production was sensitive to disruption of Golgi-ER trafficking. The detection of this oligosaccharide was unaffected by the overexpression of EDEM1 or cytosolic mannosidase, both of which increased the production of previously characterised cytosolically localised FOS. The lumenal FOS identified are therefore distinct in their production and regulation compared to FOS produced by the conventional route of misfolded glycoproteins directly removed from the ER. The production of such lumenal FOS is indicative of a novel degradative route for cellular glycoproteins that may exist under certain conditions.


Asunto(s)
Retículo Endoplásmico/fisiología , Glicoproteínas/fisiología , Oligosacáridos/análisis , Pliegue de Proteína , Proteolisis , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , Animales , Western Blotting , Células CHO , Bovinos , Línea Celular , Cromatografía Líquida de Alta Presión , Cricetinae , Cricetulus , Digitonina , Fluorescencia , Glicoproteínas/metabolismo , Inhibidores de Glicósido Hidrolasas , Aparato de Golgi/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
10.
J Am Chem Soc ; 135(26): 9723-32, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23745692

RESUMEN

Biologically active conformations of the IgG1 Fc homodimer are maintained by multiple hydrophobic interactions between the protein surface and the N-glycan. The Fc glycan modulates biological effector functions, including antibody-dependent cellular cytotoxicity (ADCC) which is mediated in part through the activatory Fc receptor, FcγRIIIA. Consistent with previous reports, we found that site-directed mutations disrupting the protein-carbohydrate interface (F241A, F243A, V262E, and V264E) increased galactosylation and sialylation of the Fc and, concomitantly, reduced the affinity for FcγRIIIA. We rationalized this effect by crystallographic analysis of the IgG1 Fc F241A mutant, determined here to a resolution of 1.9 Å, which revealed localized destabilization of this glycan-protein interface. Given that sialylation of Fc glycans decreases ADCC, one explanation for the effect of these mutants on FcγRIIIA binding is their increased sialylation. However, a glycan-engineered IgG1 with hypergalactosylated and hypersialylated glycans exhibited unchanged binding affinity to FcγRIIIA. Moreover, when we expressed these mutants as a chemically uniform (Man5GlcNAc2) glycoform, the individual effect of each mutation on FcγRIIIA affinity was preserved. This effect was broadly recapitulated for other Fc receptors (FcγRI, FcγRIIA, FcγRIIB, and FcγRIIIB). These data indicate that destabilization of the glycan-protein interactions, rather than increased galactosylation and sialylation, modifies the Fc conformation(s) relevant for FcγR binding. Engineering of the protein-carbohydrate interface thus provides an independent parameter in the engineering of Fc effector functions and a route to the synthesis of new classes of Fc domain with novel combinations of affinities for activatory and inhibitory Fc receptors.


Asunto(s)
Anticuerpos Monoclonales/química , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/química , Polisacáridos/química , Ingeniería de Proteínas , Cristalografía por Rayos X , Interacciones Hidrofóbicas e Hidrofílicas , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Modelos Moleculares
11.
J Org Chem ; 78(15): 7380-97, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23688199

RESUMEN

The Ho crossed aldol condensation provides access to a series of carbon branched iminosugars as exemplified by the synthesis of enantiomeric pairs of isoDMDP, isoDGDP, and isoDAB, allowing comparison of their biological activities with three linear isomeric natural products DMDP, DGDP, and DAB and their enantiomers. L-IsoDMDP [(2S,3S,4R)-2,4-bis(hydroxymethyl)pyrrolidine-3,4-diol], prepared in 11 steps in an overall yield of 45% from d-lyxonolactone, is a potent specific competitive inhibitor of gut disaccharidases [K(i) 0.081 µM for rat intestinal maltase] and is more effective in the suppression of hyperglycaemia in a maltose loading test than miglitol, a drug presently used in the treatment of late onset diabetes. The partial rescue of the defective F508del-CFTR function in CF-KM4 cells by L-isoDMDP is compared with miglustat and isoLAB in an approach to the treatment of cystic fibrosis.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Inhibidores de la Angiogénesis/farmacología , Productos Biológicos/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores de Glicósido Hidrolasas , Iminoazúcares/farmacología , 1-Desoxinojirimicina/farmacología , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Productos Biológicos/síntesis química , Productos Biológicos/química , Relación Dosis-Respuesta a Droga , Iminoazúcares/síntesis química , Iminoazúcares/química , Conformación Molecular , Estereoisomerismo , Relación Estructura-Actividad , alfa-Glucosidasas/metabolismo
12.
Science ; 382(6671): eabo7201, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37943932

RESUMEN

We report the results of the COVID Moonshot, a fully open-science, crowdsourced, and structure-enabled drug discovery campaign targeting the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease. We discovered a noncovalent, nonpeptidic inhibitor scaffold with lead-like properties that is differentiated from current main protease inhibitors. Our approach leveraged crowdsourcing, machine learning, exascale molecular simulations, and high-throughput structural biology and chemistry. We generated a detailed map of the structural plasticity of the SARS-CoV-2 main protease, extensive structure-activity relationships for multiple chemotypes, and a wealth of biochemical activity data. All compound designs (>18,000 designs), crystallographic data (>490 ligand-bound x-ray structures), assay data (>10,000 measurements), and synthesized molecules (>2400 compounds) for this campaign were shared rapidly and openly, creating a rich, open, and intellectual property-free knowledge base for future anticoronavirus drug discovery.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Proteasas 3C de Coronavirus , Inhibidores de Proteasa de Coronavirus , Descubrimiento de Drogas , SARS-CoV-2 , Humanos , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Simulación del Acoplamiento Molecular , Inhibidores de Proteasa de Coronavirus/síntesis química , Inhibidores de Proteasa de Coronavirus/química , Inhibidores de Proteasa de Coronavirus/farmacología , Relación Estructura-Actividad , Cristalografía por Rayos X
13.
Glycobiology ; 22(10): 1282-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22641772

RESUMEN

Removal of α-glucose residues from nascent glycoproteins in the early secretory pathway is a requirement for further N-glycan maturation. Although deglucosylation is a stepwise process mediated by endoplasmic reticulum-associated glucosidases I and II for most glycoproteins, Golgi endo-α-mannosidase provides a backup mechanism for glycoprotein deglucosylation. Although conserved in mammals, in certain cell lines, endomannosidase activity in vitro appears to differ from its activity in cells following glucosidase inhibition. Here, we show that in bovine cells this is explained by restricted substrate specificity allowing processing of Glc(1)Man(7)GlcNAc(1/2) and Glc(1)Man(5)GlcNAc(1/2) but not fully glucosylated glycans that build up when glucosidases are inhibited. Our data further demonstrate that such specificity is determined genetically rather than post-translationally. We also demonstrate that the bovine endomannosidase is transcriptionally upregulated by comparison with glucosidase II in Madin-Darby bovine kidney cells and speculate that this is to compensate for the reduced catalytic activity as measured in the recombinant form of the enzyme.


Asunto(s)
Manosidasas/metabolismo , Polisacáridos/metabolismo , Animales , Biocatálisis , Bovinos , Línea Celular , Glicosilación , Manosidasas/antagonistas & inhibidores , Manosidasas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
14.
Bioorg Med Chem ; 20(2): 641-9, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20971647

RESUMEN

Noeuromycin is a highly potent albeit unstable glycosidase inhibitor due to its hemiaminal function. While stable D-gluco-like analogs have been reported, no data are available for D-manno-like structures. A series of tri- and tetrahydroxylated seven-membered iminosugars displaying either a D-manno-or a L-gulo-like configuration, were synthesized from methyl α-D-mannopyranoside using a reductive amination-mediated ring expansion as the key step. Screening towards a range of commercial glycosidases demonstrated their potency as competitive glycosidase inhibitors while cellular assay showed selective albeit weak glycoprotein processing mannosidase inactivation.


Asunto(s)
Azepinas/química , Inhibidores Enzimáticos/síntesis química , Glucosamina/análogos & derivados , Glicósido Hidrolasas/antagonistas & inhibidores , Manosa/química , Azepinas/síntesis química , Inhibidores Enzimáticos/química , Glucosamina/síntesis química , Glucosamina/química , Glicósido Hidrolasas/metabolismo , Hidroxilación
15.
Biochem J ; 438(1): 133-42, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21585340

RESUMEN

During quality control in the ER (endoplasmic reticulum), nascent glycoproteins are deglucosylated by ER glucosidases I and II. In the post-ER compartments, glycoprotein endo-α-mannosidase provides an alternative route for deglucosylation. Previous evidence suggests that endomannosidase non-selectively deglucosylates glycoproteins that escape quality control in the ER, facilitating secretion of aberrantly folded as well as normal glycoproteins. In the present study, we employed FOS (free oligosaccharides) released from degrading glycoproteins as biomarkers of ERAD (ER-associated degradation), allowing us to gain a global rather than single protein-centred view of ERAD. Glucosidase inhibition was used to discriminate between glucosidase- and endomannosidase-mediated ERAD pathways. Endomannosidase expression was manipulated in CHO (Chinese-hamster ovary)-K1 cells, naturally lacking a functional version of the enzyme, and HEK (human embryonic kidney)-293T cells. Endomannosidase was shown to decrease the levels of total FOS, suggesting decreased rates of ERAD. However, following pharmacological inhibition of ER glucosidases I and II, endomannosidase expression resulted in a partial switch between glucosylated FOS, released from ER-confined glycoproteins, to deglucosylated FOS, released from endomannosidase-processed glycoproteins transported from the Golgi/ERGIC (ER/Golgi intermediate compartment) to the ER. Using this approach, we have identified a previously unknown pathway of glycoprotein flow, undetectable by the commonly employed methods, in which secretory cargo is targeted back to the ER after being processed by endomannosidase.


Asunto(s)
Retículo Endoplásmico/metabolismo , Glicoproteínas/metabolismo , Manosidasas/metabolismo , Oligosacáridos/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Western Blotting , Células CHO , Cricetinae , Cricetulus , Técnica del Anticuerpo Fluorescente , Glicosilación , Aparato de Golgi/metabolismo , Humanos , Mananos , Manosidasas/genética , Transporte de Proteínas
16.
Antiviral Res ; 199: 105269, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35227758

RESUMEN

Dendritic cells (DCs) are important targets for dengue virus (DENV) infection and play a significant role in the early immune response. Antiviral effects of iminosugars against DENV in primary cells have been demonstrated previously in monocyte-derived macrophages (MDMΦs). Given the important role played by DCs in innate immune defense against DENV, the antiviral effects of three deoxynojirimycin (DNJ) derivatives (NN-DNJ, EOO-DNJ and 2THO-DNJ) and a deoxygalactonojirimycin (DGJ) negative control were evaluated in DENV-infected primary human monocyte-derived immature DCs (imDCs). DNJ- but not DGJ-derivatives elicited antiviral activity in DENV-infected imDCs, similar to that observed in MDMΦs. The DNJ-derivatives inhibited DENV secretion in a dose-dependent manner. Endoplasmic reticulum (ER) α-glucosidase I inhibition by DNJ-derived iminosugars, at concentrations of 3.16 µM, correlated with a reduction in the specific infectivity of virions that were still secreted, as well as a reduction in DENV-induced tumour necrosis factor alpha secretion. This suggests iminosugar-mediated ER α-glucosidase I inhibition may give rise to further benefits during DENV infection, beyond the reduction in viral secretion associated with ER α-glucosidase II inhibition.


Asunto(s)
Virus del Dengue , Dengue , 1-Desoxinojirimicina/farmacología , Antivirales/farmacología , Antivirales/uso terapéutico , Células Dendríticas , Dengue/tratamiento farmacológico , Retículo Endoplásmico , Humanos , Macrófagos
17.
Biochem Soc Trans ; 39(1): 393-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21265811

RESUMEN

Robust assays for the isolation and characterization of urinary FOS (free oligosaccharides) have been developed to screen patients for altered protein and/or lipid glycosylation. A FOS analysis can therefore identify potential biomarkers for hepatocellular carcinoma, since variations in glycosylation as a result of tumorigenecity should be detectable in the FOS of patients. HCC (hepatocellular carcinoma) accounts for 80-90% of all liver cancers. It occurs more often in men than women and occurs mostly in people 50-60 years old. The disease is more common in parts of Africa and Asia than in North or South America and Europe. Using a combination of solid-phase extraction techniques and affinity chromatography, followed by separation of urinary FOS by NP (normal phase)-HPLC and HIAX (hydrophilic interaction and anion-exchange)-HPLC, more than 200 different species have been identified in patient samples. The high incidence of small sialylated oligosaccharides in HCC patients suggests that pro-inflammatory markers may be detected as early indicators of disease progression. In addition, the methods developed here to isolate and analyse excreted glycoprotein- and glycosphingolipid-bound oligosaccharides have been used to characterize changes in metabolic processes that underlie a number of human genetic disorders. The ability to predict disease status in microlitre amounts of readily available non-invasive urine samples indicates that rapid methods for screening can be developed.


Asunto(s)
Biomarcadores/orina , Polisacáridos/orina , Biomarcadores/química , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/orina , Cromatografía Líquida de Alta Presión/métodos , Progresión de la Enfermedad , Glicosilación , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/orina , Polisacáridos/química
18.
Bioorg Med Chem Lett ; 21(18): 5219-23, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21840710

RESUMEN

Valienol-derived allylic C-1 bromides have been used as carbaglycosyl donors for α-xylo configured valienamine pseudodisaccharide synthesis. We synthesised valienamine analogues of the Glc(α1→3)Glc and Glc(α1→3)Man disaccharides representing the linkages cleaved by α-Glucosidase II in N-glycan biosynthesis. These (N1→3)-linked pseudodisaccharides were found to have some α-Glucosidase II inhibitory activity, while two other (N1→6)-linked valienamine pseudodisaccharides failed to inhibit the enzyme.


Asunto(s)
Ciclohexenos/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores de Glicósido Hidrolasas , Hexosaminas/farmacología , Polisacáridos/biosíntesis , Biocatálisis , Técnicas de Química Sintética , Ciclohexenos/síntesis química , Ciclohexenos/química , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Hexosaminas/síntesis química , Hexosaminas/química , Conformación Molecular , Polisacáridos/química , Estereoisomerismo , Relación Estructura-Actividad , alfa-Glucosidasas/metabolismo
19.
Org Biomol Chem ; 9(15): 5373-88, 2011 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-21512716

RESUMEN

A series of neoglycoconjugates derived from deoxynojirimycin has been prepared by click connection with functionalised adamantanes. They have been assayed as glycosidase inhibitors, as inhibitors of the glycoenzymes relevant to the treatment of Gaucher disease, as well as correctors of the defective ion-transport protein involved in cystic fibrosis. We have demonstrated that it is possible to selectively either strongly inhibit ER-α-glucosidases and ceramide glucosyltransferase or restore the activity of CFTR in CF-KM4 cells by varying the length of the alkyl chain linking DNJ and adamantane.


Asunto(s)
1-Desoxinojirimicina/química , Antivirales/química , Inhibidores Enzimáticos/química , Glicoconjugados/química , Animales , Línea Celular , Cromatografía Líquida de Alta Presión , Química Clic , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células HL-60 , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Ratas , Bibliotecas de Moléculas Pequeñas/química
20.
Tetrahedron Lett ; 52(2): 219-223, 2011 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-21157573

RESUMEN

The syntheses of 4-C-Me-DAB [1,4-dideoxy-1,4-imino-4-C-methyl-d-arabinitol] from l-erythronolactone and of 4-C-Me-LAB [from d-erythronolactone] require only a single acetonide protecting group. The effect of pH on the NMR spectra of 4-C-Me-DAB [pK(a) of the salt around 8.4] is discussed and illustrates the need for care in analysis of both coupling constants and chemical shift. 4-C-Me-DAB (for rat intestinal sucrase K(i) 0.89 µM, IC(50) 0.41 µM) is a competitive - whereas 4-C-Me-LAB (for rat intestinal sucrase K(i) 0.95 µM, IC(50) 0.66 µM) is a non-competitive - specific and potent α-glucosidase inhibitor. A rationale for the α-glucosidase inhibition by DAB, LAB, 4-C-Me-DAB, 4-C-Me-LAB, and isoDAB - but not isoLAB - is provided. Both are inhibitors of endoplasmic reticulum (ER) resident α-glucosidase I and II.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA