Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 14(1): 948, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36804936

RESUMEN

Small molecule inhibitors of glycosylation enzymes are valuable tools for dissecting glycan functions and potential drug candidates. Screening for inhibitors of glycosyltransferases are mainly performed by in vitro enzyme assays with difficulties moving candidates to cells and animals. Here, we circumvent this by employing a cell-based screening assay using glycoengineered cells expressing tailored reporter glycoproteins. We focused on GalNAc-type O-glycosylation and selected the GalNAc-T11 isoenzyme that selectively glycosylates endocytic low-density lipoprotein receptor (LDLR)-related proteins as targets. Our screen of a limited small molecule compound library did not identify selective inhibitors of GalNAc-T11, however, we identify two compounds that broadly inhibited Golgi-localized glycosylation processes. These compounds mediate the reversible fragmentation of the Golgi system without affecting secretion. We demonstrate how these inhibitors can be used to manipulate glycosylation in cells to induce expression of truncated O-glycans and augment binding of cancer-specific Tn-glycoprotein antibodies and to inhibit expression of heparan sulfate and binding and infection of SARS-CoV-2.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Glicosilación , SARS-CoV-2/metabolismo , Glicoproteínas/metabolismo , Polisacáridos/metabolismo
2.
Vet Res Commun ; 47(2): 615-629, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36229725

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus 2 is the causative agent of Coronavirus Disease 2019 in humans. Among domestic animals, cats are more susceptible to SARS-CoV-2 than dogs. The detection of anti-SARS-CoV-2 antibodies in seemingly healthy cats and/or infected cats which are in close contact with infected humans has been described. The presence of animals that tested positive by serology or molecular techniques could represent a potential transmission pathway of SARS-CoV-2 that can spill over into urban wildlife. This study analyses the seroprevalence variation of SARS-CoV-2 in stray cats from different waves of outbreaks in a geographical area where previous seroepidemiological information of SARS-CoV-2 was available and investigate if SARS-CoV-2-seropositive cats were exposed to other co-infections causing an immunosuppressive status and/or a chronic disease that could lead to a SARS-CoV-2 susceptibility. For this purpose, a total of 254 stray cats from Zaragoza (Spain) were included. This analysis was carried out by the enzyme-linked immunosorbent assay using the receptor binding domain of Spike antigen and confirmed by serum virus neutralization assay. The presence of co-infections including Toxoplasma gondii, Leishmania infantum, Dirofilaria immitis, feline calicivirus, feline herpesvirus type 1, feline leukemia virus and feline immunodeficiency virus, was evaluated using different serological methods. A seropositivity of 1.57% was observed for SARS-CoV-2 including the presence of neutralizing antibodies in three cats. None of the seropositive to SARS-CoV-2 cats were positive to feline coronavirus, however, four SARS-CoV-2-seropositive cats were also seropositive to other pathogens such as L. infantum, D. immitis and FIV (n = 1), L. infantum and D. immitis (n = 1) and L. infantum alone (n = 1).Considering other pathogens, a seroprevalence of 16.54% was detected for L. infantum, 30.31% for D. immitis, 13.78%, for T. gondii, 83.86% for feline calicivirus, 42.52% for feline herpesvirus type 1, 3.15% for FeLV and 7.87% for FIV.Our findings suggest that the epidemiological role of stray cats in SARS-CoV-2 transmission is scarce, and there is no increase in seropositivity during the different waves of COVID-19 outbreaks in this group of animals. Further epidemiological surveillances are necessary to determine the risk that other animals might possess even though stray cats do not seem to play a role in transmission.


Asunto(s)
COVID-19 , Enfermedades de los Gatos , Coinfección , Dirofilaria immitis , Enfermedades de los Perros , Virus de la Inmunodeficiencia Felina , Humanos , Gatos , Animales , Perros , Enfermedades de los Gatos/epidemiología , SARS-CoV-2 , Coinfección/epidemiología , Coinfección/veterinaria , España/epidemiología , Estudios Seroepidemiológicos , Estudios Transversales , COVID-19/epidemiología , COVID-19/veterinaria , Virus de la Leucemia Felina , Brotes de Enfermedades , Enfermedades de los Perros/epidemiología
3.
Glycobiology ; 30(11): 910-922, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-32304323

RESUMEN

A family of polypeptide GalNAc-transferases (GalNAc-Ts) initiates mucin-type O-glycosylation, transferring GalNAc onto hydroxyl groups of Ser and Thr residues of target substrates. The 20 GalNAc-T isoenzymes in humans are classified into nine subfamilies according to sequence similarity. GalNAc-Ts select their sites of glycosylation based on weak and overlapping peptide sequence motifs, as well prior substrate O-GalNAc glycosylation at sites both remote (long-range) and neighboring (short-range) the acceptor. Together, these preferences vary among GalNAc-Ts imparting each isoenzyme with its own unique specificity. Studies on the first identified GalNAc-Ts showed Thr acceptors were preferred over Ser acceptors; however studies comparing Thr vs. Ser glycosylation across the GalNAc-T family are lacking. Using a series of identical random peptide substrates, with single Thr or Ser acceptor sites, we determined the rate differences (Thr/Ser rate ratio) between Thr and Ser substrate glycosylation for 12 isoenzymes (representing 7 GalNAc-T subfamilies). These Thr/Ser rate ratios varied across subfamilies, ranging from ~2 to ~18 (for GalNAc-T4/GalNAc-T12 and GalNAc-T3/GalNAc-T6, respectively), while nearly identical Thr/Ser rate ratios were observed for isoenzymes within subfamilies. Furthermore, the Thr/Ser rate ratios did not appreciably vary over a series of fixed sequence substrates of different relative activities, suggesting the ratio is a constant for each isoenzyme against single acceptor substrates. Finally, based on GalNAc-T structures, the different Thr/Ser rate ratios likely reflect differences in the strengths of the Thr acceptor methyl group binding to the active site pocket. With this work, another activity that further differentiates substrate specificity among the GalNAc-Ts has been identified.


Asunto(s)
Mucinas/metabolismo , N-Acetilgalactosaminiltransferasas/metabolismo , Serina/metabolismo , Treonina/metabolismo , Glicosilación , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Mucinas/química , N-Acetilgalactosaminiltransferasas/química , Serina/química , Treonina/química , Polipéptido N-Acetilgalactosaminiltransferasa
4.
Curr Opin Struct Biol ; 56: 87-96, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30703750

RESUMEN

Mucin-type O-glycosylation is a post-translational modification (PTM) that is predicted to occur in more than the 80% of the proteins that pass through the Golgi apparatus. This PTM is initiated by a family of polypeptide GalNAc-transferases (GalNAc-Ts) that modify Ser and Thr residues of proteins through the addition of a GalNAc moiety. These enzymes are type II membrane proteins that consist of a Golgi luminal catalytic domain connected by a flexible linker to a ricin type lectin domain. Together, both domains account for the different glycosylation preferences observed among isoenzymes. Although it is well accepted that most of the family members share some degree of redundancy toward their protein and glycoprotein substrates, it has been recently found that several GalNAc-Ts also possess activity toward specific targets. Despite the high similarity between isoenzymes, structural differences have recently been reported that are key to understanding the molecular basis of both their redundancy and specificity. The present review focuses on the molecular aspects of the protein substrate recognition and the different glycosylation preferences of these enzymes, which in turn will serve as a roadmap to the rational design of specific modulators of mucin-type O-glycosylation.


Asunto(s)
N-Acetilgalactosaminiltransferasas/metabolismo , Dominio Catalítico , Glicopéptidos/metabolismo , Glicosilación , Humanos , N-Acetilgalactosaminiltransferasas/química , Especificidad por Sustrato , Polipéptido N-Acetilgalactosaminiltransferasa
5.
ACS Cent Sci ; 4(9): 1274-1290, 2018 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-30276263

RESUMEN

Mucin-type O-glycosylation is initiated by a family of polypeptide GalNAc-transferases (GalNAc-Ts) which are type-II transmembrane proteins that contain Golgi luminal catalytic and lectin domains that are connected by a flexible linker. Several GalNAc-Ts, including GalNAc-T4, show both long-range and short-range prior glycosylation specificity, governed by their lectin and catalytic domains, respectively. While the mechanism of the lectin-domain-dependent glycosylation is well-known, the molecular basis for the catalytic-domain-dependent glycosylation of glycopeptides is unclear. Herein, we report the crystal structure of GalNAc-T4 bound to the diglycopeptide GAT*GAGAGAGT*TPGPG (containing two α-GalNAc glycosylated Thr (T*), the PXP motif and a "naked" Thr acceptor site) that describes its catalytic domain glycopeptide GalNAc binding site. Kinetic studies of wild-type and GalNAc binding site mutant enzymes show the lectin domain GalNAc binding activity dominates over the catalytic domain GalNAc binding activity and that these activities can be independently eliminated. Surprisingly, a flexible loop protruding from the lectin domain was found essential for the optimal activity of the catalytic domain. This work provides the first structural basis for the short-range glycosylation preferences of a GalNAc-T.

6.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 8): 443-450, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30084393

RESUMEN

Protein O-fucosylation is an important post-translational modification (PTM) found in cysteine-rich repeats in proteins. Protein O-fucosyltransferases 1 and 2 (PoFUT1 and PoFUT2) are the enzymes responsible for this PTM and selectively glycosylate specific residues in epidermal growth factor-like (EGF) repeats and thrombospondin type I repeats (TSRs), respectively. Within the past six years, crystal structures of both enzymes have been reported, revealing important information on how they recognize protein substrates and achieve catalysis. Here, the structural information available today is summarized and how PoFUT1 and PoFUT2 employ different catalytic mechanisms is discussed.


Asunto(s)
Fucosiltransferasas/química , Fucosiltransferasas/metabolismo , Animales , Humanos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Galactósido 2-alfa-L-Fucosiltransferasa
7.
J Biol Chem ; 293(19): 7408-7422, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29559555

RESUMEN

The low-density lipoprotein receptor (LDLR) and related receptors are important for the transport of diverse biomolecules across cell membranes and barriers. Their functions are especially relevant for cholesterol homeostasis and diseases, including neurodegenerative and kidney disorders. Members of the LDLR-related protein family share LDLR class A (LA) repeats providing binding properties for lipoproteins and other biomolecules. We previously demonstrated that short linker regions between these LA repeats contain conserved O-glycan sites. Moreover, we found that O-glycan modifications at these sites are selectively controlled by the GalNAc-transferase isoform, GalNAc-T11. However, the effects of GalNAc-T11-mediated O-glycosylation on LDLR and related receptor localization and function are unknown. Here, we characterized O-glycosylation of LDLR-related proteins and identified conserved O-glycosylation sites in the LA linker regions of VLDLR, LRP1, and LRP2 (Megalin) from both cell lines and rat organs. Using a panel of gene-edited isogenic cell line models, we demonstrate that GalNAc-T11-mediated LDLR and VLDLR O-glycosylation is not required for transport and cell-surface expression and stability of these receptors but markedly enhances LDL and VLDL binding and uptake. Direct ELISA-based binding assays with truncated LDLR constructs revealed that O-glycosylation increased affinity for LDL by ∼5-fold. The molecular basis for this observation is currently unknown, but these findings open up new avenues for exploring the roles of LDLR-related proteins in disease.


Asunto(s)
Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Receptores de LDL/metabolismo , Acetilgalactosamina/metabolismo , Animales , Células CHO , Membrana Celular/metabolismo , Cricetulus , Drosophila , Glicosilación , Células HEK293 , Células Hep G2 , Humanos , Ligandos , Lipoproteínas/metabolismo , Polisacáridos/metabolismo , Unión Proteica , Transporte de Proteínas , Ratas , Proteínas Recombinantes/metabolismo
8.
Chemistry ; 24(33): 8382-8392, 2018 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-29601100

RESUMEN

The family of polypeptide N-acetylgalactosamine (GalNAc) transferases (GalNAc-Ts) orchestrates the initiating step of mucin-type protein O-glycosylation by transfer of GalNAc moieties to serine and threonine residues in proteins. Deficiencies and dysregulation of GalNAc-T isoenzymes are related to different diseases. Recently, it has been demonstrated that an inactive GalNAc-T2 mutant (F104S), which is not located at the active site, induces low levels of high-density lipoprotein cholesterol (HDL-C) in humans. Herein, the molecular basis for F104S mutant inactivation has been deciphered. Saturation transfer difference NMR spectroscopy experiments demonstrate that the mutation induces loss of binding to peptide substrates. Analysis of the crystal structure of the F104S mutant bound to UDP-GalNAc (UDP=uridine diphosphate), combined with molecular dynamics (MD) simulations, has revealed that the flexible loop is disordered and displays larger conformational changes in the mutant enzyme than that in the wild-type (WT) enzyme. 19 F NMR spectroscopy experiments reveal that the WT enzyme only reaches the active state in the presence of UDP-GalNAc, which provides compelling evidence that GalNAc-T2 adopts a UDP-GalNAc-dependent induced-fit mechanism. The F104S mutation precludes the enzyme from achieving the active conformation and concomitantly binding peptide substrates. This study provides new insights into the catalytic mechanism of the large family of GalNAc-Ts and how these enzymes orchestrate protein O-glycosylation.


Asunto(s)
Mucina-1/análisis , Mucina-1/química , Mucinas/química , N-Acetilgalactosaminiltransferasas/análisis , N-Acetilgalactosaminiltransferasas/química , Uridina Difosfato/química , Catálisis , Dominio Catalítico , Glicosilación , Humanos , Simulación de Dinámica Molecular , Polipéptido N-Acetilgalactosaminiltransferasa
9.
Nat Commun ; 8(1): 1959, 2017 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-29208955

RESUMEN

The polypeptide GalNAc-transferases (GalNAc-Ts), that initiate mucin-type O-glycosylation, consist of a catalytic and a lectin domain connected by a flexible linker. In addition to recognizing polypeptide sequence, the GalNAc-Ts exhibit unique long-range N- and/or C-terminal prior glycosylation (GalNAc-O-Ser/Thr) preferences modulated by the lectin domain. Here we report studies on GalNAc-T4 that reveal the origins of its unique N-terminal long-range glycopeptide specificity, which is the opposite of GalNAc-T2. The GalNAc-T4 structure bound to a monoglycopeptide shows that the GalNAc-binding site of its lectin domain is rotated relative to the homologous GalNAc-T2 structure, explaining their different long-range preferences. Kinetics and molecular dynamics simulations on several GalNAc-T2 flexible linker constructs show altered remote prior glycosylation preferences, confirming that the flexible linker dictates the rotation of the lectin domain, thus modulating the GalNAc-Ts' long-range preferences. This work for the first time provides the structural basis for the different remote prior glycosylation preferences of the GalNAc-Ts.


Asunto(s)
N-Acetilgalactosaminiltransferasas/química , N-Acetilgalactosaminiltransferasas/metabolismo , Péptidos/química , Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Quimera/genética , Clonación Molecular , Glicopéptidos/química , Glicopéptidos/metabolismo , Glicosilación , Humanos , Cinética , Lectinas/química , Lectinas/metabolismo , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , N-Acetilgalactosaminiltransferasas/genética , Unión Proteica , Conformación Proteica , Especificidad por Sustrato , Polipéptido N-Acetilgalactosaminiltransferasa
10.
Chemistry ; 22(21): 7215-24, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27071848

RESUMEN

The Leloir donors are nucleotide sugars essential for a variety of glycosyltransferases (GTs) involved in the transfer of a carbohydrate to an acceptor substrate, typically a protein or an oligosaccharide. A series of less-polar nucleotide sugar analogues derived from uridine have been prepared by replacing one phosphate unit with an alkyl chain. The methodology is based on the radical hydrophosphonylation of alkenes, which allows coupling of allyl glycosyl compounds with a phosphate unit suitable for conjugation to uridine. Two of these compounds, the GalNAc and galactose derivatives, were further tested on a model GT, such as GalNAc-T2 (an important GT widely distributed in human tissues), to probe that both compounds bound in the medium-high micromolar range. The crystal structure of GalNAc-T2 with the galactose derivative traps the enzyme in an inactive form; this suggests that compounds only containing the ß-phosphate could be efficient ligands for the enzyme. Computational studies with GalNAc-T2 corroborate these findings and provide further insights into the mechanism of the catalytic cycle of this family of enzymes.


Asunto(s)
Glicoconjugados/química , Glicoconjugados/metabolismo , Glicosiltransferasas/metabolismo , N-Acetilgalactosaminiltransferasas/metabolismo , Uridina/análogos & derivados , Uridina/metabolismo , Alquilación , Dominio Catalítico , Galactosa/análogos & derivados , Galactosa/metabolismo , Humanos , Simulación del Acoplamiento Molecular , N-Acetilgalactosaminiltransferasas/química , Conformación Proteica , Especificidad por Sustrato , Polipéptido N-Acetilgalactosaminiltransferasa
11.
Nat Commun ; 6: 6937, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25939779

RESUMEN

Protein O-glycosylation is controlled by polypeptide GalNAc-transferases (GalNAc-Ts) that uniquely feature both a catalytic and lectin domain. The underlying molecular basis of how the lectin domains of GalNAc-Ts contribute to glycopeptide specificity and catalysis remains unclear. Here we present the first crystal structures of complexes of GalNAc-T2 with glycopeptides that together with enhanced sampling molecular dynamics simulations demonstrate a cooperative mechanism by which the lectin domain enables free acceptor sites binding of glycopeptides into the catalytic domain. Atomic force microscopy and small-angle X-ray scattering experiments further reveal a dynamic conformational landscape of GalNAc-T2 and a prominent role of compact structures that are both required for efficient catalysis. Our model indicates that the activity profile of GalNAc-T2 is dictated by conformational heterogeneity and relies on a flexible linker located between the catalytic and the lectin domains. Our results also shed light on how GalNAc-Ts generate dense decoration of proteins with O-glycans.


Asunto(s)
Dominio Catalítico , Lectinas/química , N-Acetilgalactosaminiltransferasas/química , N-Acetilgalactosaminiltransferasas/metabolismo , Cristalografía por Rayos X , Glicopéptidos/química , Glicopéptidos/metabolismo , Glicosilación , Modelos Moleculares , Simulación de Dinámica Molecular , Nucleótidos/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato , Polipéptido N-Acetilgalactosaminiltransferasa
12.
Angew Chem Int Ed Engl ; 53(31): 8206-10, 2014 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-24954443

RESUMEN

The retaining glycosyltransferase GalNAc-T2 is a member of a large family of human polypeptide GalNAc-transferases that is responsible for the post-translational modification of many cell-surface proteins. By the use of combined structural and computational approaches, we provide the first set of structural snapshots of the enzyme during the catalytic cycle and combine these with quantum-mechanics/molecular-mechanics (QM/MM) metadynamics to unravel the catalytic mechanism of this retaining enzyme at the atomic-electronic level of detail. Our study provides a detailed structural rationale for an ordered bi-bi kinetic mechanism and reveals critical aspects of substrate recognition, which dictate the specificity for acceptor Thr versus Ser residues and enforce a front-face SN i-type reaction in which the substrate N-acetyl sugar substituent coordinates efficient glycosyl transfer.


Asunto(s)
N-Acetilgalactosaminiltransferasas/química , Conformación Proteica , Especificidad por Sustrato , Polipéptido N-Acetilgalactosaminiltransferasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA