Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 298(8): 102206, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35772493

RESUMEN

Mutations in the SLC35C1 gene encoding the Golgi GDP-fucose transporter are known to cause leukocyte adhesion deficiency II. However, improvement of fucosylation in leukocyte adhesion deficiency II patients treated with exogenous fucose suggests the existence of an SLC35C1-independent route of GDP-fucose transport, which remains a mystery. To investigate this phenomenon, we developed and characterized a human cell-based model deficient in SLC35C1 activity. The resulting cells were cultured in the presence/absence of exogenous fucose and mannose, followed by examination of fucosylation potential and nucleotide sugar levels. We found that cells displayed low but detectable levels of fucosylation in the absence of SLC35C1. Strikingly, we show that defects in fucosylation were almost completely reversed upon treatment with millimolar concentrations of fucose. Furthermore, we show that even if fucose was supplemented at nanomolar concentrations, it was still incorporated into glycans by these knockout cells. We also found that the SLC35C1-independent transport preferentially utilized GDP-fucose from the salvage pathway over the de novo biogenesis pathway as a source of this substrate. Taken together, our results imply that the Golgi systems of GDP-fucose transport discriminate between substrate pools obtained from different metabolic pathways, which suggests a functional connection between nucleotide sugar transporters and nucleotide sugar synthases.


Asunto(s)
Fucosa , Guanosina Difosfato Fucosa , Síndrome de Deficiencia de Adhesión del Leucocito/terapia , Fucosa/metabolismo , Aparato de Golgi/metabolismo , Guanosina Difosfato Fucosa/metabolismo , Humanos , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Polisacáridos/metabolismo
2.
J Biol Chem ; 296: 100299, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33460651

RESUMEN

The human Gb3/CD77 synthase, encoded by the A4GALT gene, is an unusually promiscuous glycosyltransferase. It synthesizes the Galα1→4Gal linkage on two different glycosphingolipids (GSLs), producing globotriaosylceramide (Gb3, CD77, Pk) and the P1 antigen. Gb3 is the major receptor for Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli. A single amino acid substitution (p.Q211E) ramps up the enzyme's promiscuity, rendering it able to attach Gal both to another Gal residue and to GalNAc, giving rise to NOR1 and NOR2 GSLs. Human Gb3/CD77 synthase was long believed to transfer Gal only to GSL acceptors, therefore its GSL products were, by default, considered the only human Stx receptors. Here, using soluble, recombinant human Gb3/CD77 synthase and p.Q211E mutein, we demonstrate that both enzymes can synthesize the P1 glycotope (terminal Galα1→4Galß1→4GlcNAc-R) on a complex type N-glycan and a synthetic N-glycoprotein (saposin D). Moreover, by transfection of CHO-Lec2 cells with vectors encoding human Gb3/CD77 synthase and its p.Q211E mutein, we demonstrate that both enzymes produce P1 glycotopes on N-glycoproteins, with the mutein exhibiting elevated activity. These P1-terminated N-glycoproteins are recognized by Stx1 but not Stx2 B subunits. Finally, cytotoxicity assays show that Stx1 can use P1 N-glycoproteins produced in CHO-Lec2 cells as functional receptors. We conclude that Stx1 can recognize and use P1 N-glycoproteins in addition to its canonical GSL receptors to enter and kill the cells, while Stx2 can use GSLs only. Collectively, these results may have important implications for our understanding of the Shiga toxin pathology.


Asunto(s)
Galactosiltransferasas/química , Globósidos/química , Toxina Shiga I/química , Trihexosilceramidas/química , Acetilgalactosamina/química , Acetilgalactosamina/metabolismo , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Animales , Sitios de Unión , Células CHO , Secuencia de Carbohidratos , Cricetulus , Escherichia coli Enterohemorrágica/química , Escherichia coli Enterohemorrágica/patogenicidad , Galactosa/química , Galactosa/metabolismo , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Expresión Génica , Globósidos/biosíntesis , Globósidos/metabolismo , Glucosa/química , Glucosa/metabolismo , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Toxina Shiga I/metabolismo , Toxina Shiga II/química , Toxina Shiga II/metabolismo , Trihexosilceramidas/biosíntesis
3.
Biochem Biophys Res Commun ; 635: 46-51, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36257191

RESUMEN

The sialylation of glycoconjugates is performed by a variety of sialyltransferases using CMP-sialic acid (CMP-Sia) as a substrate. Sialylation requires the translocation of CMP-Sia across the Golgi membranes. This function has been assigned to SLC35A1, the only CMP-Sia transporter identified to date. Mutations in the SLC35A1 gene cause a subtype of congenital disorder of glycosylation (CDG). Over the past several years, heterologous complexes formed in the Golgi membrane by some SLC35A subfamily members and functionally related glycosyltransferases have been reported. However, to date no interaction between SLC35A1 and a sialyltransferase has been identified. In this study we attempted to clarify the role of SLC35A1 in α2,3 sialylation of N-glycans. We showed that SLC35A1 associates with ST3Gal4, the main α2,3-sialyltransferase acting on N-glycans. This phenomenon is compromised by the E196K (but not T156R) mutation in the SLC35A1 gene. We also demonstrated that the E196K mutant is less efficient in restoring N-glycan sialylation upon expression in the SLC35A1 knockout cells. On the basis of our findings, we propose that the interaction between SLC35A1 and ST3Gal4 may be important for proper sialylation.


Asunto(s)
Ácido N-Acetilneuramínico Citidina Monofosfato , Sialiltransferasas , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Ácido N-Acetilneuramínico Citidina Monofosfato/metabolismo , Glicosilación , Mutación , Polisacáridos
4.
Int J Mol Sci ; 23(15)2022 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-35955785

RESUMEN

Nucleotide sugars (NSs) serve as substrates for glycosylation reactions. The majority of these compounds are synthesized in the cytoplasm, whereas glycosylation occurs in the endoplasmic reticulum (ER) and Golgi lumens, where catalytic domains of glycosyltransferases (GTs) are located. Therefore, translocation of NS across the organelle membranes is a prerequisite. This process is thought to be mediated by a group of multi-transmembrane proteins from the SLC35 family, i.e., nucleotide sugar transporters (NSTs). Despite many years of research, some uncertainties/inconsistencies related with the mechanisms of NS transport and the substrate specificities of NSTs remain. Here we present a comprehensive review of the NS import into the mammalian Golgi, which consists of three major parts. In the first part, we provide a historical view of the experimental approaches used to study NS transport and evaluate the most important achievements. The second part summarizes various aspects of knowledge concerning NSTs, ranging from subcellular localization up to the pathologies related with their defective function. In the third part, we present the outcomes of our research performed using mammalian cell-based models and discuss its relevance in relation to the general context.


Asunto(s)
Aparato de Golgi , Nucleótidos , Azúcares , Animales , Transporte Biológico , Glicosilación , Aparato de Golgi/metabolismo , Mamíferos/metabolismo , Nucleótidos/metabolismo , Azúcares/metabolismo
5.
J Biol Chem ; 295(48): 16445-16463, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-32938718

RESUMEN

Nucleotide sugar transporters, encoded by the SLC35 gene family, deliver nucleotide sugars throughout the cell for various glycosyltransferase-catalyzed glycosylation reactions. GlcNAc, in the form of UDP-GlcNAc, and galactose, as UDP-Gal, are delivered into the Golgi apparatus by SLC35A3 and SLC35A2 transporters, respectively. However, although the UDP-Gal transporting activity of SLC35A2 has been clearly demonstrated, UDP-GlcNAc delivery by SLC35A3 is not fully understood. Therefore, we analyzed a panel of CHO, HEK293T, and HepG2 cell lines including WT cells, SLC35A2 knockouts, SLC35A3 knockouts, and double-knockout cells. Cells lacking SLC35A2 displayed significant changes in N- and O-glycan synthesis. However, in SLC35A3-knockout CHO cells, only limited changes were observed; GlcNAc was still incorporated into N-glycans, but complex type N-glycan branching was impaired, although UDP-GlcNAc transport into Golgi vesicles was not decreased. In SLC35A3-knockout HEK293T cells, UDP-GlcNAc transport was significantly decreased but not completely abolished. However, N-glycan branching was not impaired in these cells. In CHO and HEK293T cells, the effect of SLC35A3 deficiency on N-glycan branching was potentiated in the absence of SLC35A2. Moreover, in SLC35A3-knockout HEK293T and HepG2 cells, GlcNAc was still incorporated into O-glycans. However, in the case of HepG2 cells, no qualitative changes in N-glycans between WT and SLC35A3 knockout cells nor between SLC35A2 knockout and double-knockout cells were observed. These findings suggest that SLC35A3 may not be the primary UDP-GlcNAc transporter and/or different mechanisms of UDP-GlcNAc transport into the Golgi apparatus may exist.


Asunto(s)
Glicosiltransferasas/metabolismo , Aparato de Golgi/metabolismo , Proteínas de Transporte de Nucleótidos/metabolismo , Polisacáridos/biosíntesis , Animales , Células CHO , Cricetulus , Técnicas de Silenciamiento del Gen , Glicosiltransferasas/genética , Aparato de Golgi/genética , Células HEK293 , Células Hep G2 , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas de Transporte de Nucleótidos/genética , Polisacáridos/genética
6.
Glycobiology ; 31(9): 1145-1162, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-33978735

RESUMEN

N-glycosylation is a ubiquitous posttranslational modification that may influence folding, subcellular localization, secretion, solubility and oligomerization of proteins. In this study, we examined the effects of N-glycans on the activity of human Gb3/CD77 synthase, which catalyzes the synthesis of glycosphingolipids with terminal Galα1→4Gal (Gb3 and the P1 antigen) and Galα1→4GalNAc disaccharides (the NOR antigen). The human Gb3/CD77 synthase contains two occupied N-glycosylation sites at positions N121 and N203. Intriguingly, we found that while the N-glycan at N203 is essential for activity and correct subcellular localization, the N-glycan at N121 is dispensable and its absence did not reduce, but, surprisingly, even increased the activity of the enzyme. The fully N-glycosylated human Gb3/CD77 synthase and its glycoform missing the N121 glycan correctly localized in the Golgi, whereas a glycoform without the N203 site partially mislocalized in the endoplasmic reticulum. A double mutein missing both N-glycans was inactive and accumulated in the endoplasmic reticulum. Our results suggest that the decreased specific activity of human Gb3/CD77 synthase glycovariants resulted from their improper subcellular localization and, to a smaller degree, a decrease in enzyme solubility. Taken together, our findings show that the two N-glycans of human Gb3/CD77 synthase have opposing effects on its properties, revealing a dual nature of N-glycosylation and potentially a novel regulatory mechanism controlling the biological activity of proteins.


Asunto(s)
Galactosiltransferasas , Glicoesfingolípidos , Galactosiltransferasas/metabolismo , Glicosilación , Humanos , Polisacáridos , Trihexosilceramidas
7.
Biochem J ; 477(2): 381-405, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31899475

RESUMEN

As part of the infective process, Porphyromonas gingivalis must acquire heme which is indispensable for life and enables the microorganism to survive and multiply at the infection site. This oral pathogenic bacterium uses a newly discovered novel hmu heme uptake system with a leading role played by the HmuY hemophore-like protein, responsible for acquiring heme and increasing virulence of this periodontopathogen. We demonstrated that Prevotella intermedia produces two HmuY homologs, termed PinO and PinA. Both proteins were produced at higher mRNA and protein levels when the bacterium grew under low-iron/heme conditions. PinO and PinA bound heme, but preferentially under reducing conditions, and in a manner different from that of the P. gingivalis HmuY. The analysis of the three-dimensional structures confirmed differences between apo-PinO and apo-HmuY, mainly in the fold forming the heme-binding pocket. Instead of two histidine residues coordinating heme iron in P. gingivalis HmuY, PinO and PinA could use one methionine residue to fulfill this function, with potential support of additional methionine residue/s. The P. intermedia proteins sequestered heme only from the host albumin-heme complex under reducing conditions. Our findings suggest that HmuY-like family might comprise proteins subjected during evolution to significant diversification, resulting in different heme coordination modes. The newer data presented in this manuscript on HmuY homologs produced by P. intermedia sheds more light on the novel mechanism of heme uptake, could be helpful in discovering their biological function, and in developing novel therapeutic approaches.


Asunto(s)
Hemo/genética , Hemoproteínas/genética , Periodontitis/genética , Prevotella intermedia/genética , Regulación Bacteriana de la Expresión Génica/genética , Hemo/química , Hemoproteínas/química , Humanos , Hierro/metabolismo , Periodontitis/microbiología , Periodontitis/patología , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/patogenicidad , Prevotella intermedia/patogenicidad , ARN Mensajero/genética , Homología de Secuencia de Aminoácido
8.
Anal Biochem ; 593: 113599, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32004544

RESUMEN

Split luciferase complementation assay is one of the approaches enabling identification and analysis of protein-protein interactions in vivo. The NanoBiT technology is the most recent improvement of this strategy. Nucleotide sugar transporters and glycosyltransferases of the Golgi apparatus are the key players in glycosylation. Here we demonstrate the applicability of the NanoBiT system for studying homooligomerization of these proteins. We also report and discuss a novel heterologous interaction between UDP-galactose transporter and beta-1,4-galactosyltransferase 1.


Asunto(s)
Mediciones Luminiscentes/métodos , Proteínas de Transporte de Monosacáridos/metabolismo , N-Acetil-Lactosamina Sintasa/metabolismo , Nanotecnología/métodos , Secuencia de Aminoácidos , Animales , Transporte Biológico , Células CHO , Cricetulus , Aparato de Golgi/metabolismo , Células HEK293 , Humanos , Unión Proteica
9.
Glycoconj J ; 37(5): 577-588, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32827291

RESUMEN

ß1,4-galactosyltransferase 4 (B4GalT4) is one of seven B4GalTs that belong to CAZy glycosyltransferase family 7 and transfer galactose to growing sugar moieties of proteins, glycolipids, glycosaminoglycans as well as single sugar for lactose synthesis. Herein, we identify two asparagine-linked glycosylation sites in B4GalT4. We found that mutation of one site (Asn220) had greater impact on enzymatic activity while another (Asn335) on Golgi localization and presence of N-glycans at both sites is required for production of stable and enzymatically active protein and its secretion. Additionally, we confirm B4GalT4 involvement in synthesis of keratan sulfate (KS) by generating A375 B4GalT4 knock-out cell lines that show drastic decrease in the amount of KS proteoglycans and no significant structural changes in N- and O-glycans. We show that KS decrease in A375 cells deficient in B4GalT4 activity can be rescued by overproduction of either partially or fully glycosylated B4GalT4 but not with N-glycan-depleted B4GalT4 version.


Asunto(s)
Galactosiltransferasas/genética , Glicosaminoglicanos/genética , Aparato de Golgi/genética , Polisacáridos/genética , Línea Celular , Galactosa/genética , Galactosiltransferasas/química , Técnicas de Inactivación de Genes , Glicosaminoglicanos/química , Glicosilación , Humanos , Sulfato de Queratano/química , Polisacáridos/metabolismo
10.
Cell Mol Life Sci ; 76(9): 1821-1832, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30737517

RESUMEN

Branching and processing of N-glycans in the medial-Golgi rely both on the transport of the donor UDP-N-acetylglucosamine (UDP-GlcNAc) to the Golgi lumen by the SLC35A3 nucleotide sugar transporter (NST) as well as on the addition of the GlcNAc residue to terminal mannoses in nascent N-glycans by several linkage-specific N-acetyl-glucosaminyltransferases (MGAT1-MGAT5). Previous data indicate that the MGATs and NSTs both form higher order assemblies in the Golgi membranes. Here, we investigate their specific and mutual interactions using high-throughput FRET- and BiFC-based interaction screens. We show that MGAT1, MGAT2, MGAT3, MGAT4B (but not MGAT5) and Golgi alpha-mannosidase IIX (MAN2A2) form several distinct molecular assemblies with each other and that the MAN2A2 acts as a central hub for the interactions. Similar assemblies were also detected between the NSTs SLC35A2, SLC35A3, and SLC35A4. Using in vivo BiFC-based FRET interaction screens, we also identified novel ternary complexes between the MGATs themselves or between the MGATs and the NSTs. These findings suggest that the MGATs and the NSTs self-assemble into multi-enzyme/multi-transporter complexes in the Golgi membranes in vivo to facilitate efficient synthesis of complex N-glycans.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Complejos Multienzimáticos/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Proteínas de Transporte de Nucleótidos/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Polisacáridos/metabolismo , Uridina Difosfato N-Acetilglucosamina/metabolismo , alfa-Manosidasa/metabolismo
11.
Int J Mol Sci ; 22(1)2020 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-33396746

RESUMEN

Congenital disorders of glycosylation (CDG) are a group of rare genetic and metabolic diseases caused by alterations in glycosylation pathways. Five patients bearing CDG-causing mutations in the SLC35A1 gene encoding the CMP-sialic acid transporter (CST) have been reported to date. In this study we examined how specific mutations in the SLC35A1 gene affect the protein's properties in two previously described SLC35A1-CDG cases: one caused by a substitution (Q101H) and another involving a compound heterozygous mutation (T156R/E196K). The effects of single mutations and the combination of T156R and E196K mutations on the CST's functionality was examined separately in CST-deficient HEK293T cells. As shown by microscopic studies, none of the CDG-causing mutations affected the protein's proper localization in the Golgi apparatus. Cellular glycophenotypes were characterized using lectins, structural assignment of N- and O-glycans and analysis of glycolipids. Single Q101H, T156R and E196K mutants were able to partially restore sialylation in CST-deficient cells, and the deleterious effect of a single T156R or E196K mutation on the CST functionality was strongly enhanced upon their combination. We also revealed differences in the ability of CST variants to form dimers. The results of this study improve our understanding of the molecular background of SLC35A1-CDG cases.


Asunto(s)
Mutación , Proteínas de Transporte de Nucleótidos/genética , Proteínas de Transporte de Nucleótidos/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Simportadores/genética , Simportadores/metabolismo , Sistemas CRISPR-Cas , Membrana Celular/metabolismo , Cromatografía Líquida de Alta Presión , Citidina Monofosfato/metabolismo , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Glicoconjugados/metabolismo , Glicosilación , Células HEK293 , Humanos , Lectinas/metabolismo
12.
Hum Mutat ; 40(7): 908-925, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30817854

RESUMEN

Pathogenic de novo variants in the X-linked gene SLC35A2 encoding the major Golgi-localized UDP-galactose transporter required for proper protein and lipid glycosylation cause a rare type of congenital disorder of glycosylation known as SLC35A2-congenital disorders of glycosylation (CDG; formerly CDG-IIm). To date, 29 unique de novo variants from 32 unrelated individuals have been described in the literature. The majority of affected individuals are primarily characterized by varying degrees of neurological impairments with or without skeletal abnormalities. Surprisingly, most affected individuals do not show abnormalities in serum transferrin N-glycosylation, a common biomarker for most types of CDG. Here we present data characterizing 30 individuals and add 26 new variants, the single largest study involving SLC35A2-CDG. The great majority of these individuals had normal transferrin glycosylation. In addition, expanding the molecular and clinical spectrum of this rare disorder, we developed a robust and reliable biochemical assay to assess SLC35A2-dependent UDP-galactose transport activity in primary fibroblasts. Finally, we show that transport activity is directly correlated to the ratio of wild-type to mutant alleles in fibroblasts from affected individuals.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Uridina Difosfato Galactosa/metabolismo , Animales , Biopsia , Células CHO , Células Cultivadas , Trastornos Congénitos de Glicosilación/metabolismo , Trastornos Congénitos de Glicosilación/patología , Cricetulus , Femenino , Humanos , Masculino , Mutación
13.
BMC Microbiol ; 19(1): 127, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31185896

RESUMEN

BACKGROUND: Porphyromonas gingivalis is considered a keystone pathogen responsible for chronic periodontitis. Although several virulence factors produced by this bacterium are quite well characterized, very little is known about regulatory mechanisms that allow different strains of P. gingivalis to efficiently survive in the hostile environment of the oral cavity, a typical habitat characterized by low iron and heme concentrations. The aim of this study was to characterize P. gingivalis Fur homolog (PgFur) in terms of its role in production of virulence factors in more (A7436) and less (ATCC 33277) virulent strains. RESULTS: Expression of a pgfur depends on the growth phase and iron/heme concentration. To better understand the role played by the PgFur protein in P. gingivalis virulence under low- and high-iron/heme conditions, a pgfur-deficient ATCC 33277 strain (TO16) was constructed and its phenotype compared with that of a pgfur A7436-derived mutant strain (TO6). In contrast to the TO6 strain, the TO16 strain did not differ in the growth rate and hemolytic activity compared with the ATCC 33277 strain. However, both mutant strains were more sensitive to oxidative stress and they demonstrated changes in the production of lysine- (Kgp) and arginine-specific (Rgp) gingipains. In contrast to the wild-type strains, TO6 and TO16 mutant strains produced larger amounts of HmuY protein under high iron/heme conditions. We also demonstrated differences in production of glycoconjugates between the A7436 and ATCC 33277 strains and we found evidence that PgFur protein might regulate glycosylation process. Moreover, we revealed that PgFur protein plays a role in interactions with other periodontopathogens and is important for P. gingivalis infection of THP-1-derived macrophages and survival inside the cells. Deletion of the pgfur gene influences expression of many transcription factors, including two not yet characterized transcription factors from the Crp/Fnr family. We also observed lower expression of the CRISPR/Cas genes. CONCLUSIONS: We show here for the first time that inactivation of the pgfur gene exerts a different influence on the phenotype of the A7436 and ATCC 33277 strains. Our findings further support the hypothesis that PgFur regulates expression of genes encoding surface virulence factors and/or genes involved in their maturation.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Metaloproteínas/genética , Porphyromonas gingivalis/crecimiento & desarrollo , Factores de Virulencia/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Infecciones por Bacteroidaceae/microbiología , Periodontitis Crónica/microbiología , Regulación Bacteriana de la Expresión Génica , Glicosilación , Humanos , Hierro/metabolismo , Metaloproteínas/metabolismo , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo , Porphyromonas gingivalis/metabolismo , Porphyromonas gingivalis/patogenicidad , Células THP-1 , Factores de Virulencia/metabolismo
14.
Int J Mol Sci ; 20(2)2019 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-30641943

RESUMEN

Solute carrier family 35 member A5 (SLC35A5) is a member of the SLC35A protein subfamily comprising nucleotide sugar transporters. However, the function of SLC35A5 is yet to be experimentally determined. In this study, we inactivated the SLC35A5 gene in the HepG2 cell line to study a potential role of this protein in glycosylation. Introduced modification affected neither N- nor O-glycans. There was also no influence of the gene knock-out on glycolipid synthesis. However, inactivation of the SLC35A5 gene caused a slight increase in the level of chondroitin sulfate proteoglycans. Moreover, inactivation of the SLC35A5 gene resulted in the decrease of the uridine diphosphate (UDP)-glucuronic acid, UDP-N-acetylglucosamine, and UDP-N-acetylgalactosamine Golgi uptake, with no influence on the UDP-galactose transport activity. Further studies demonstrated that SLC35A5 localized exclusively to the Golgi apparatus. Careful insight into the protein sequence revealed that the C-terminus of this protein is extremely acidic and contains distinctive motifs, namely DXEE, DXD, and DXXD. Our studies show that the C-terminus is directed toward the cytosol. We also demonstrated that SLC35A5 formed homomers, as well as heteromers with other members of the SLC35A protein subfamily. In conclusion, the SLC35A5 protein might be a Golgi-resident multiprotein complex member engaged in nucleotide sugar transport.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas de Transporte de Nucleótidos/genética , Proteínas de Transporte de Nucleótidos/metabolismo , Proteínas Transportadoras de Solutos/genética , Proteínas Transportadoras de Solutos/metabolismo , Azúcares de Uridina Difosfato/metabolismo , Secuencias de Aminoácidos , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Citosol/metabolismo , Técnicas de Inactivación de Genes , Glicosilación , Células Hep G2 , Humanos , Proteínas de Transporte de Nucleótidos/química , Uridina Difosfato Ácido Glucurónico/metabolismo , Uridina Difosfato N-Acetilglucosamina/metabolismo
15.
Biochim Biophys Acta Mol Cell Res ; 1864(5): 825-838, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28167211

RESUMEN

SLC35A4 has been classified in the SLC35A subfamily based on amino acid sequence homology. Most of the proteins belonging to the SLC35 family act as transporters of nucleotide sugars. In this study, the subcellular localization of endogenous SLC35A4 was determined via immunofluorescence staining, and it was demonstrated that SLC35A4 localizes mainly to the Golgi apparatus. In silico topology prediction suggests that SLC35A4 has an uneven number of transmembrane domains and its N-terminus is directed towards the Golgi lumen. However, an experimental assay refuted this prediction: SLC35A4 has an even number of transmembrane regions with both termini facing the cytosol. In vivo interaction analysis using the FLIM-FRET approach revealed that SLC35A4 neither forms homomers nor associates with other members of the SLC35A subfamily except SLC35A5. Additional assays demonstrated that endogenous SLC35A4 is 10 to 40nm proximal to SLC35A2 and SLC35A3. To determine SLC35A4 function SLC35A4 knock-out cells were generated with the CRISPR-Cas9 approach. Although no significant changes in glycosylation were observed, the introduced mutation influenced the subcellular distribution of the SLC35A2/SLC35A3 complexes. Additional FLIM-FRET experiments revealed that overexpression of SLC35A4-BFP together with SLC35A3 and the SLC35A2-Golgi splice variant negatively affects the interaction between the two latter proteins. The results presented here strongly indicate a modulatory role for SLC35A4 in intracellular trafficking of SLC35A2/SLC35A3 complexes.


Asunto(s)
Proteínas de Transporte de Monosacáridos/fisiología , Proteínas de Transporte de Nucleótidos/fisiología , Secuencia de Aminoácidos , Animales , Transporte Biológico/genética , Células COS , Metabolismo de los Hidratos de Carbono/genética , Línea Celular Tumoral , Chlorocebus aethiops , Perros , Células HEK293 , Células Hep G2 , Humanos , Células de Riñón Canino Madin Darby , Proteínas de Transporte de Monosacáridos/química , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Nucleótidos/química , Proteínas de Transporte de Nucleótidos/genética , Homología de Secuencia de Aminoácido
16.
Anaerobe ; 44: 27-33, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28104533

RESUMEN

Porphyromonas gingivalis is a major etiologic agent and a key pathogen responsible for the development and progression of chronic periodontitis. Controlling the number of periodontal pathogens is one of the primary actions for maintaining oral health; therefore, active compounds with a capacity to exert antimicrobial activity have received considerable attention as they may represent potential new therapeutic agents for the treatment of chronic periodontitis. Heterocyclic compounds possessing 1,2,4- or 1,2,3-triazoles are known for several biological activities, including antibacterial properties. Among them are stable hemiaminals which can be obtained in reaction between nitrobenzaldehyde derivatives and 4-amino-1,2,4-triazole or 4-amino-3,5-dimethyl-1,2,4-triazole. In this study, we selected two relatively stable hemiaminals: (2,4-dinitrophenyl)(4H-1,2,4-triazole-4-ylamino)methanol (24DNTAM) and (2,4-dinitrophenyl)(4H-3,5-dimethyl-1,2,4-triazole-4-ylamino)methanol (24DNDMTAM). Both compounds showed promising anti-P. gingivalis activity, higher against ATCC 33277 strain as compared to A7436 strain. The lowest hemiaminal concentration inhibiting visible planktonic bacterial growth under high-iron/heme conditions was ∼0.06 mg/ml, and the lowest hemiaminal concentration showing killing of bacteria was ∼0.25 mg/ml. Antimicrobial activity was also observed against P. gingivalis grown on blood agar plates. Slightly higher antimicrobial activity of both compounds was observed when P. gingivalis was grown in co-cultures with epithelial HeLa cells under low-iron/heme conditions, which mimic those occurring in vivo. 24DNTAM was more effective against P. gingivalis, but exhibited higher cytotoxic activity against epithelial and red blood cells, as compared with 24DNDMTAM. We conclude that both hemiaminals might originate a novel group of biologically important molecules.


Asunto(s)
Antibacterianos/farmacología , Porphyromonas gingivalis/efectos de los fármacos , Triazoles/farmacología , Antibacterianos/síntesis química , Técnicas de Cocultivo , Células Epiteliales/microbiología , Células Epiteliales/fisiología , Células HeLa , Humanos , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Modelos Moleculares , Estructura Molecular , Porphyromonas gingivalis/crecimiento & desarrollo , Triazoles/síntesis química
17.
J Biol Chem ; 290(25): 15475-15486, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-25944901

RESUMEN

UDP-galactose transporter (UGT; SLC35A2) and UDP-N-acetylglucosamine transporter (NGT; SLC35A3) form heterologous complexes in the Golgi membrane. NGT occurs in close proximity to mannosyl (α-1,6-)-glycoprotein ß-1,6-N-acetylglucosaminyltransferase (Mgat5). In this study we analyzed whether NGT and both splice variants of UGT (UGT1 and UGT2) are able to interact with four different mannoside acetylglucosaminyltransferases (Mgat1, Mgat2, Mgat4B, and Mgat5). Using an in situ proximity ligation assay, we found that all examined glycosyltransferases are in the vicinity of these UDP-sugar transporters both at the endogenous level and upon overexpression. This observation was confirmed via the FLIM-FRET approach for both NGT and UGT1 complexes with Mgats. This study reports for the first time close proximity between endogenous nucleotide sugar transporters and glycosyltransferases. We also observed that among all analyzed Mgats, only Mgat4B occurs in close proximity to UGT2, whereas the other three Mgats are more distant from UGT2, and it was only possible to visualize their vicinity using proximity ligation assay. This strongly suggests that the distance between these protein pairs is longer than 10 nm but at the same time shorter than 40 nm. This study adds to the understanding of glycosylation, one of the most important post-translational modifications, which affects the majority of macromolecules. Our research shows that complex formation between nucleotide sugar transporters and glycosyltransferases might be a more common phenomenon than previously thought.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Animales , Transporte Biológico Activo/fisiología , Línea Celular Tumoral , Perros , Transferencia Resonante de Energía de Fluorescencia , Glicosilación , Aparato de Golgi/química , Aparato de Golgi/genética , Humanos , Células de Riñón Canino Madin Darby , Proteínas de Transporte de Monosacáridos/química , Proteínas de Transporte de Monosacáridos/genética , N-Acetilglucosaminiltransferasas/química , N-Acetilglucosaminiltransferasas/genética
18.
Biochem Biophys Res Commun ; 467(4): 748-53, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26482851

RESUMEN

Porphyromonas gingivalis, the main etiologic agent and key pathogen responsible for initiation and progression of chronic periodontitis, is a haem auxotroph, and the uptake of this compound is essential for its survival and the ability to establish an infection. The aim of this study was to examine the role of a hemophore-like HmuY protein in P. gingivalis growth and infection of macrophages. Inactivation of the hmuY gene caused reduced P. gingivalis growth in vitro in the presence of serum as a heme sole source, as well as in vivo co-cultures with THP-1-derived macrophages. This resulted in diminished invasion efficiency of macrophages by live bacteria lacking functional hmuY gene. Both features were partially restored after addition of the purified HmuY protein, which was internalized when added either together with the hmuY mutant strain or alone to macrophage cultures. We conclude that HmuY is an important virulence factor of P. gingivalis for infection of macrophages in a heme-limited host environment.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hemo/metabolismo , Macrófagos/microbiología , Porphyromonas gingivalis/patogenicidad , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Infecciones por Bacteroidaceae/microbiología , Línea Celular/microbiología , Interacciones Huésped-Patógeno , Humanos , Mutación , Porphyromonas gingivalis/genética , Factores de Virulencia/genética
19.
J Biol Chem ; 288(30): 21850-60, 2013 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-23766508

RESUMEN

SLC35A3 is considered the main UDP-N-acetylglucosamine transporter (NGT) in mammals. Detailed analysis of NGT is restricted because mammalian mutant cells defective in this activity have not been isolated. Therefore, using the siRNA approach, we developed and characterized several NGT-deficient mammalian cell lines. CHO, CHO-Lec8, and HeLa cells deficient in NGT activity displayed a decrease in the amount of highly branched tri- and tetraantennary N-glycans, whereas monoantennary and diantennary ones remained unchanged or even were accumulated. Silencing the expression of NGT in Madin-Darby canine kidney II cells resulted in a dramatic decrease in the keratan sulfate content, whereas no changes in biosynthesis of heparan sulfate were observed. We also demonstrated for the first time close proximity between NGT and mannosyl (α-1,6-)-glycoprotein ß-1,6-N-acetylglucosaminyltransferase (Mgat5) in the Golgi membrane. We conclude that NGT may be important for the biosynthesis of highly branched, multiantennary complex N-glycans and keratan sulfate. We hypothesize that NGT may specifically supply ß-1,3-N-acetylglucosaminyl-transferase 7 (ß3GnT7), Mgat5, and possibly mannosyl (α-1,3-)-glycoprotein ß-1,4-N-acetylglucosaminyltransferase (Mgat4) with UDP-GlcNAc.


Asunto(s)
Sulfato de Queratano/biosíntesis , Proteínas de Transporte de Membrana/metabolismo , Polisacáridos/biosíntesis , Interferencia de ARN , Animales , Secuencia de Bases , Transporte Biológico , Células CHO , Línea Celular , Línea Celular Tumoral , Cricetinae , Cricetulus , Perros , Transferencia Resonante de Energía de Fluorescencia , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Proteínas de Transporte de Membrana/genética , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Análisis de Secuencia de ADN , Azúcares de Uridina Difosfato/metabolismo
20.
Biochem Biophys Res Commun ; 454(4): 486-92, 2014 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-25451267

RESUMEN

UDP-galactose transporter (UGT) and UDP-N-acetylglucosamine transporter (NGT) form heterologous complexes in the Golgi apparatus (GA) membrane. We aimed to identify UGT region responsible for galactosylation of N-glycans. Chimeric proteins composed of human UGT and either NGT or CMP-sialic acid transporter (CST) localized to the GA, and all but UGT/CST chimera corrected galactosylation defect in UGT-deficient cell lines, although at different efficiency. Importantly, short N-terminal region composed of 35 N-terminal amino-acid residues of UGT was crucial for galactosylation of N-glycans. The remaining molecule must be derived from NGT not CST, confirming that the role played by UGT and NGT is coupled.


Asunto(s)
Galactosa/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Polisacáridos/química , Polisacáridos/metabolismo , Animales , Células CHO , Cricetulus , Perros , Glicosilación , Humanos , Células de Riñón Canino Madin Darby
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA