Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
J Biotechnol ; 379: 53-64, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38070779

RESUMO

The baculovirus-insect cell expression system allows addition of O-fucose to EGF-like domains of glycoproteins, following the action of the protein O-fucosyltransferase 1 named POFUT1. In this study, recombinant Spodoptera frugiperda POFUT1 from baculovirus-infected Sf9 cells was compared to recombinant Mus musculus POFUT1 produced by CHO cells. Contrary to recombinant murine POFUT1 carrying two hybrid and/or complex type N-glycans, Spodoptera frugiperda POFUT1 exhibited paucimannose N-glycans, at least on its highly evolutionary conserved across Metazoa NRT site. The abilities of both recombinant enzymes to add in vitro O -fucose to EGF-like domains of three different recombinant mammalian glycoproteins were then explored. In vitro POFUT1-mediated O-fucosylation experiments, followed by click chemistry and blot analyses, showed that Spodoptera frugiperda POFUT1 was able to add O-fucose to mouse NOTCH1 EGF-like 26 and WIF1 EGF-like 3 domains, similarly to the murine counterpart. As proved by mass spectrometry, full-length human WNT Inhibitor Factor 1 expressed by Sf9 cells was also modified with O-fucose. However, Spodoptera frugiperda POFUT1 was unable to modify the single EGF-like domain of mouse PAMR1 with O-fucose, contrary to murine POFUT1. Absence of orthologous proteins such as PAMR1 in insects may explain the enzyme's difficulty in adding O-fucose to a domain that it never encounters naturally.


Assuntos
Fucosiltransferases , Proteínas Recombinantes , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Spodoptera/enzimologia , Spodoptera/genética , Spodoptera/metabolismo , Fucosiltransferases/química , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Humanos , Animais , Camundongos , Células CHO , Cricetulus , Células Sf9 , Glicosilação , Sequência Consenso , Fucose/metabolismo , Domínios Proteicos
2.
Nat Chem Biol ; 19(8): 1022-1030, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37202521

RESUMO

Mammalian cell surface and secreted glycoproteins exhibit remarkable glycan structural diversity that contributes to numerous physiological and pathogenic interactions. Terminal glycan structures include Lewis antigens synthesized by a collection of α1,3/4-fucosyltransferases (CAZy GT10 family). At present, the only available crystallographic structure of a GT10 member is that of the Helicobacter pylori α1,3-fucosyltransferase, but mammalian GT10 fucosyltransferases are distinct in sequence and substrate specificity compared with the bacterial enzyme. Here, we determined crystal structures of human FUT9, an α1,3-fucosyltransferase that generates Lewisx and Lewisy antigens, in complex with GDP, acceptor glycans, and as a FUT9-donor analog-acceptor Michaelis complex. The structures reveal substrate specificity determinants and allow prediction of a catalytic model supported by kinetic analyses of numerous active site mutants. Comparisons with other GT10 fucosyltransferases and GT-B fold glycosyltransferases provide evidence for modular evolution of donor- and acceptor-binding sites and specificity for Lewis antigen synthesis among mammalian GT10 fucosyltransferases.


Assuntos
Fucosiltransferases , Glicosiltransferases , Animais , Humanos , Fucosiltransferases/genética , Fucosiltransferases/química , Fucosiltransferases/metabolismo , Antígenos do Grupo Sanguíneo de Lewis , Polissacarídeos/metabolismo , Mamíferos
3.
Drug Discov Today ; 28(1): 103394, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36223858

RESUMO

Fucosyltransferases (FUTs) have significant roles in various pathophysiological events. Their high expression is a signature of malignant cell transformation, contributing to many abnormal events during cancer development, such as uncontrolled cell proliferation, tumor cell invasion, angiogenesis, metastasis, immune evasion, and therapy resistance. Therefore, FUTs have evolved as an attractive therapeutic target for treating solid cancers, and many substrate analogs have been discovered with potential as FUT inhibitors for cancer therapy. Meanwhile, the development of FUT protein structures represents a significant advance in the design of FUT inhibitors with nonsubstrate structures. In this review, we summarize the role of FUTs in cancers, the resolved protein crystal structures and progress in the development of FUT inhibitors as cancer therapeutics.


Assuntos
Fucosiltransferases , Neoplasias , Humanos , Fucosiltransferases/química , Fucosiltransferases/metabolismo , Glicosilação , Neoplasias/tratamento farmacológico , Proliferação de Células
4.
PLoS One ; 16(10): e0257623, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34648519

RESUMO

Fucosyltransferase 2 (FUT2) catalyzes the biosynthesis of A, B, and H antigens and other important glycans, such as (Sialyl Lewisx) sLex, and (Sialyl Lewisy) sLey. The production of these glycans is increased in various cancers, hence to design and develop specific inhibitors of FUT2 is a therapeutic strategy. The current study was designed to identify the inhibitors for FUT2. In silico screening of 300 synthetic compounds was performed. Molecular docking studies highlighted the interactions of ligands with critical amino acid residues, present in the active site of FUT2. The epitope mapping in ligands was performed using the STD-NMR experiments to identify the interactions between ligands, and receptor protein. Finally, we have identified 5 lead compounds 4, 5, 26, 27, and 28 that can be studied for further development as cancer therapeutic agents.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Fucosiltransferases/antagonistas & inibidores , Domínio Catalítico/efeitos dos fármacos , Fucosiltransferases/química , Fucosiltransferases/metabolismo , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Simulação de Acoplamento Molecular , Galactosídeo 2-alfa-L-Fucosiltransferase
5.
Molecules ; 26(17)2021 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-34500643

RESUMO

Mammalian cell surfaces are modified with complex arrays of glycans that play major roles in health and disease. Abnormal glycosylation is a hallmark of cancer; terminal sialic acid and fucose in particular have high levels in tumor cells, with positive implications for malignancy. Increased sialylation and fucosylation are due to the upregulation of a set of sialyltransferases (STs) and fucosyltransferases (FUTs), which are potential drug targets in cancer. In the past, several advances in glycostructural biology have been made with the determination of crystal structures of several important STs and FUTs in mammals. Additionally, how the independent evolution of STs and FUTs occurred with a limited set of global folds and the diverse modular ability of catalytic domains toward substrates has been elucidated. This review highlights advances in the understanding of the structural architecture, substrate binding interactions, and catalysis of STs and FUTs in mammals. While this general understanding is emerging, use of this information to design inhibitors of STs and FUTs will be helpful in providing further insights into their role in the manifestation of cancer and developing targeted therapeutics in cancer.


Assuntos
Fucosiltransferases/química , Fucosiltransferases/metabolismo , Mamíferos/metabolismo , Sialiltransferases/química , Sialiltransferases/metabolismo , Animais , Catálise , Domínio Catalítico/fisiologia , Glicosilação , Humanos
6.
Biochem J ; 478(8): 1571-1583, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33734311

RESUMO

The α1,6-fucosyltransferase, FUT8, is the sole enzyme catalyzing the core-fucosylation of N-glycoproteins in mammalian systems. Previous studies using free N-glycans as acceptor substrates indicated that a terminal ß1,2-GlcNAc moiety on the Man-α1,3-Man arm of N-glycan substrates is required for efficient FUT8-catalyzed core-fucosylation. In contrast, we recently demonstrated that, in a proper protein context, FUT8 could also fucosylate Man5GlcNAc2 without a GlcNAc at the non-reducing end. We describe here a further study of the substrate specificity of FUT8 using a range of N-glycans containing different aglycones. We found that FUT8 could fucosylate most of high-mannose and complex-type N-glycans, including highly branched N-glycans from chicken ovalbumin, when the aglycone moiety is modified with a 9-fluorenylmethyloxycarbonyl (Fmoc) moiety or in a suitable peptide/protein context, even if they lack the terminal GlcNAc moiety on the Man-α1,3-Man arm. FUT8 could also fucosylate paucimannose structures when they are on glycoprotein substrates. Such core-fucosylated paucimannosylation is a prominent feature of lysosomal proteins of human neutrophils and several types of cancers. We also found that sialylation of N-glycans significantly reduced their activity as a substrate of FUT8. Kinetic analysis demonstrated that Fmoc aglycone modification could either improve the turnover rate or decrease the KM value depending on the nature of the substrates, thus significantly enhancing the overall efficiency of FUT8 catalyzed fucosylation. Our results indicate that an appropriate aglycone context of N-glycans could significantly broaden the acceptor substrate specificity of FUT8 beyond what has previously been thought.


Assuntos
Eritropoetina/metabolismo , Fucose/metabolismo , Fucosiltransferases/metabolismo , Glicoproteínas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Manose/metabolismo , Polissacarídeos/metabolismo , Animais , Sequência de Carboidratos , Galinhas , Eritropoetina/química , Eritropoetina/genética , Fluorenos/química , Fucose/química , Fucosiltransferases/química , Fucosiltransferases/genética , Expressão Gênica , Glicoproteínas/química , Glicoproteínas/genética , Glicosilação , Fator Estimulador de Colônias de Granulócitos e Macrófagos/química , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Células HEK293 , Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/genética , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/genética , HIV-1/metabolismo , Humanos , Cinética , Manose/química , Ovalbumina/química , Ovalbumina/genética , Ovalbumina/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Polissacarídeos/química , Especificidade por Substrato
7.
J Biol Chem ; 295(50): 17027-17045, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33004438

RESUMO

Mammalian Asn-linked glycans are extensively processed as they transit the secretory pathway to generate diverse glycans on cell surface and secreted glycoproteins. Additional modification of the glycan core by α-1,6-fucose addition to the innermost GlcNAc residue (core fucosylation) is catalyzed by an α-1,6-fucosyltransferase (FUT8). The importance of core fucosylation can be seen in the complex pathological phenotypes of FUT8 null mice, which display defects in cellular signaling, development, and subsequent neonatal lethality. Elevated core fucosylation has also been identified in several human cancers. However, the structural basis for FUT8 substrate specificity remains unknown.Here, using various crystal structures of FUT8 in complex with a donor substrate analog, and with four distinct glycan acceptors, we identify the molecular basis for FUT8 specificity and activity. The ordering of three active site loops corresponds to an increased occupancy for bound GDP, suggesting an induced-fit folding of the donor-binding subsite. Structures of the various acceptor complexes were compared with kinetic data on FUT8 active site mutants and with specificity data from a library of glycan acceptors to reveal how binding site complementarity and steric hindrance can tune substrate affinity. The FUT8 structure was also compared with other known fucosyltransferases to identify conserved and divergent structural features for donor and acceptor recognition and catalysis. These data provide insights into the evolution of modular templates for donor and acceptor recognition among GT-B fold glycosyltransferases in the synthesis of diverse glycan structures in biological systems.


Assuntos
Fucosiltransferases/química , Dobramento de Proteína , Cristalografia por Raios X , Células HEK293 , Humanos , Domínios Proteicos , Homologia Estrutural de Proteína , Especificidade por Substrato
8.
Int J Mol Sci ; 21(17)2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32825463

RESUMO

Fucosylated glycans critically regulate the physiological functions of proteins and cells. Alterations in levels of fucosylated glycans are associated with various diseases. For detection and functional modulation of fucosylated glycans, chemical biology approaches using fucose (Fuc) analogs are useful. However, little is known about how efficiently each unnatural Fuc analog is utilized by enzymes in the biosynthetic pathway of fucosylated glycans. We show here that three clickable Fuc analogs with similar but distinct structures labeled cellular glycans with different efficiency and protein specificity. For instance, 6-alkynyl (Alk)-Fuc modified O-Fuc glycans much more efficiently than 7-Alk-Fuc. The level of GDP-6-Alk-Fuc produced in cells was also higher than that of GDP-7-Alk-Fuc. Comprehensive in vitro fucosyltransferase assays revealed that 7-Alk-Fuc is commonly tolerated by most fucosyltransferases. Surprisingly, both protein O-fucosyltransferases (POFUTs) could transfer all Fuc analogs in vitro, likely because POFUT structures have a larger space around their Fuc binding sites. These findings demonstrate that labeling and detection of fucosylated glycans with Fuc analogs depend on multiple cellular steps, including conversion to GDP form, transport into the ER or Golgi, and utilization by each fucosyltransferase, providing insights into design of novel sugar analogs for specific detection of target glycans or inhibition of their functions.


Assuntos
Fucose/análogos & derivados , Fucose/química , Fucosiltransferases/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Sítios de Ligação , Biotinilação , Química Click , Fucose/metabolismo , Fucosiltransferases/química , Fucosiltransferases/genética , Glicosilação , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Células HEK293 , Humanos , Polissacarídeos/química , Polissacarídeos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombospondina 1/genética , Trombospondina 1/metabolismo
9.
J Biol Chem ; 295(19): 6677-6688, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32220931

RESUMO

Fucosylation of the innermost GlcNAc of N-glycans by fucosyltransferase 8 (FUT8) is an important step in the maturation of complex and hybrid N-glycans. This simple modification can dramatically affect the activities and half-lives of glycoproteins, effects that are relevant to understanding the invasiveness of some cancers, development of mAb therapeutics, and the etiology of a congenital glycosylation disorder. The acceptor substrate preferences of FUT8 are well-characterized and provide a framework for understanding N-glycan maturation in the Golgi; however, the structural basis of these substrate preferences and the mechanism through which catalysis is achieved remain unknown. Here we describe several structures of mouse and human FUT8 in the apo state and in complex with GDP, a mimic of the donor substrate, and with a glycopeptide acceptor substrate at 1.80-2.50 Å resolution. These structures provide insights into a unique conformational change associated with donor substrate binding, common strategies employed by fucosyltransferases to coordinate GDP, features that define acceptor substrate preferences, and a likely mechanism for enzyme catalysis. Together with molecular dynamics simulations, the structures also revealed how FUT8 dimerization plays an important role in defining the acceptor substrate-binding site. Collectively, this information significantly builds on our understanding of the core fucosylation process.


Assuntos
Fucosiltransferases/química , Guanosina Difosfato/química , Simulação de Dinâmica Molecular , Animais , Sítios de Ligação , Catálise , Cristalografia por Raios X , Humanos , Camundongos
10.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 8): 443-450, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30084393

RESUMO

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.


Assuntos
Fucosiltransferases/química , Fucosiltransferases/metabolismo , Animais , Humanos , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Galactosídeo 2-alfa-L-Fucosiltransferase
11.
J Am Chem Soc ; 139(28): 9431-9434, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28678517

RESUMO

The core fucosylation of N-glycans on glycoproteins is catalyzed by fucosyltransferase 8 (FUT8) in mammalian cells and is involved in various biological functions, such as protein function, cancer progression, and postnatal development. The substrate specificity of FUT8 toward bi-antennary N-glycans has been reported, but it is unclear with regard to tri-antennary and tetra-antennary glycans. Here, we examined the specificity and activity of human FUT8 toward tri- and tetra-antennary N-glycans in the forms of glycopeptides. We found that the tri-antennary glycan [A3(2,4,2) type] terminated with N-acetylglucosamine (GlcNAc), which is generated by N-acetylglucosaminyltransferase (GnT)-IV, is a good substrate for FUT8, but the A3(2,2,6) type of tri-antennary glycan, generated by GnT-V, is not a substrate for FUT8. We also observed that core fucosylation reduced the activity of GnT-IV toward the bi-antennary glycan. Examining the correlation between the types of N-glycans and the expression levels of FUT8, GnT-IV, and GnT-V in cells revealed that these glycosyltransferases, particularly GnT-IV, play important roles in directing the branching and core fucosylation of N-glycans in vivo. This study thus provides insights into the interplay among FUT8, GnT-IV, and GnT-V in N-linked glycosylation during the assembly of glycoproteins.


Assuntos
Fucose/metabolismo , Fucosiltransferases/metabolismo , Glicoproteínas/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Polissacarídeos/metabolismo , Biocatálise , Fucose/química , Fucosiltransferases/química , Glicoproteínas/química , Glicosilação , Humanos , N-Acetilglucosaminiltransferases/química , Polissacarídeos/química , Especificidade por Substrato
12.
Glycobiology ; 27(7): 601-618, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28430973

RESUMO

Fucose is a 6-deoxy hexose in the l-configuration found in a large variety of different organisms. In mammals, fucose is incorporated into N-glycans, O-glycans and glycolipids by 13 fucosyltransferases, all of which utilize the nucleotide-charged form, GDP-fucose, to modify targets. Three of the fucosyltransferases, FUT8, FUT12/POFUT1 and FUT13/POFUT2, are essential for proper development in mice. Fucose modifications have also been implicated in many other biological functions including immunity and cancer. Congenital mutations of a Golgi apparatus localized GDP-fucose transporter causes leukocyte adhesion deficiency type II, which results in severe developmental and immune deficiencies, highlighting the important role fucose plays in these processes. Additionally, changes in levels of fucosylated proteins have proven as useful tools for determining cancer diagnosis and prognosis. Chemically modified fucose analogs can be used to alter many of these fucose dependent processes or as tools to better understand them. In this review, we summarize the known roles of fucose in mammalian physiology and pathophysiology. Additionally, we discuss recent therapeutic advances for cancer and other diseases that are a direct result of our improved understanding of the role that fucose plays in these systems.


Assuntos
Fucose/metabolismo , Fucosiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Fucosiltransferases/química , Fucosiltransferases/genética , Humanos , Doenças do Sistema Imunitário/genética , Doenças do Sistema Imunitário/metabolismo , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Mutação , Neoplasias/genética , Neoplasias/metabolismo
13.
Glycobiology ; 27(8): 777-786, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28334865

RESUMO

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


Assuntos
Fucosiltransferases/química , Fucosiltransferases/genética , Hiperpigmentação/genética , Mutação , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/genética , Dermatopatias Genéticas/genética , Dermatopatias Papuloescamosas/genética , Carcinogênese/genética , Carcinogênese/metabolismo , Fucosiltransferases/deficiência , Fucosiltransferases/metabolismo , Humanos , Hiperpigmentação/metabolismo , Ligantes , Conformação Proteica , Dermatopatias Genéticas/metabolismo , Dermatopatias Papuloescamosas/metabolismo
14.
Plant Signal Behav ; 10(6): e1026023, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26176901

RESUMO

Two independent studies have shown that the cell wall of pollen tubes from tobacco and tomato species contained fucosylated xyloglucan (XyG). These findings are intriguing as many reports have shown that XyG of somatic cells of these species is not fucosylated but instead is arabinosylated. In order to produce fucosylated XyG, plants must express a functional galactoside α-2-fucosyltransferase. Here, using a bioinformatics approach, we show that several candidate genes coding for XyG fucosyltransferases are present in the genome of coffee and several Solanaceae species including tomato, tobacco, potato, eggplant and pepper. BLAST and protein alignments with the 2 well-characterized XyG fucosyltransferases from Arabidopsis thaliana and Pisum sativum revealed that at least 6 proteins from different Solanaceae species and from coffee displayed the 3 conserved motifs required for XyG fucosyltransferase activity.


Assuntos
Fucosiltransferases/metabolismo , Genoma de Planta , Proteínas de Plantas/metabolismo , Solanaceae/enzimologia , Solanaceae/genética , Algoritmos , Motivos de Aminoácidos , Sequência de Aminoácidos , Coffea/enzimologia , Simulação por Computador , Fucosiltransferases/química , Dados de Sequência Molecular , Filogenia , Proteínas de Plantas/química , Estrutura Terciária de Proteína , Alinhamento de Sequência
15.
Int J Oncol ; 47(2): 701-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26094873

RESUMO

Abnormal glycosylation is catalyzed by the specific glycosyltransferases and correlates with tumor cell apoptosis. Increased fucosyltransferase IV (FUT4) is seen in many types of cancer, and manipulating FUT4 expression through specific signaling pathway inhibits cell growth and induces apoptosis. NF-κB is known playing a vital role to control cell growth and apoptosis. Ginsenoside Rg3 is an herbal medicine with strong antitumor activity through inhibiting tumor growth and promoting tumor cell death. However, whether Rg3-induced inhibition on tumor development involves reduced NF-κB signaling and FUT4 expression remains unknown. In the present study, we found that Rg3 suppressed FUT4 expression by abrogating the binding of NF-κB to FUT4 promoter through inhibiting the expression of signaling molecules of NF-κB pathway, reducing NF-κB DNA binding activity and NF-κB transcription activity. NF-κB inhibitor (Bay 11-7082) or knocking down p65 expression by p65 siRNA also led to a significant decreased FUT4 expression. In addition, Rg3 induced apoptosis by activating both extrinsic and intrinsic apoptotic pathways. Moreover, in a xenograft mouse model, Rg3 downregulated FUT4 and NF-κB/p65 expression and suppressed melanoma cell growth and induced apoptosis without any noticeable toxicity. In conclusion, Rg3 induces tumor cell apoptosis correlated with its inhibitory effect on NF-κB signaling pathway-mediated FUT4 expression. Results suggest Rg3 might be a novel therapy agent for melanoma treatment.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Fucosiltransferases/genética , Ginsenosídeos/administração & dosagem , Antígenos CD15/genética , Melanoma/tratamento farmacológico , NF-kappa B/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/farmacologia , Apoptose , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fucosiltransferases/química , Fucosiltransferases/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ginsenosídeos/farmacologia , Humanos , Antígenos CD15/química , Antígenos CD15/metabolismo , Masculino , Melanoma/genética , Melanoma/metabolismo , Camundongos , NF-kappa B/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Bioorg Med Chem Lett ; 25(12): 2501-4, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25956415

RESUMO

Two fluorescent diboronic acid compounds (6a and 6b) with a dipeptide linker were synthesized as potential sensors for cell surface saccharide Lewis X (Le(X)). Compound 6a with a dipeptide (H-Asp-Ala-) as the linker was found to selectively label CHOFUT4 cells, which express Le(x), at micromolar concentrations, while non-Le(x)-expressing control cells were not labeled.


Assuntos
Antracenos/química , Ácidos Borônicos/química , Corantes Fluorescentes/química , Oligossacarídeos/metabolismo , Animais , Antracenos/síntese química , Antracenos/metabolismo , Ácidos Borônicos/síntese química , Ácidos Borônicos/metabolismo , Células CHO , Cricetinae , Cricetulus , Dipeptídeos/química , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/metabolismo , Fucosiltransferases/química , Fucosiltransferases/metabolismo , Antígenos do Grupo Sanguíneo de Lewis , Microscopia de Fluorescência , Oligossacarídeos/genética
17.
ACS Chem Biol ; 8(8): 1830-40, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23730796

RESUMO

α1,6-Core-fucosyltransferase (FUT8) is a vital enzyme in mammalian physiological and pathophysiological processes such as tumorigenesis and progress of, among others, non-small cell lung cancer and colon carcinoma. It was also shown that therapeutic antibodies have a dramatically higher efficacy if the α1,6-fucosyl residue is absent. However, specific and potent inhibitors for FUT8 and related enzymes are lacking. Hence, it is crucial to elucidate the structural basis of acceptor binding and the catalytic mechanism. We present here the first structural model of FUT8 in complex with its acceptor and donor molecules. An unusually large acceptor, i.e., a hexasaccharide from the core of N-glycans, is required as minimal structure. Acceptor substrate binding of FUT8 is being dissected experimentally by STD NMR and SPR and theoretically by molecular dynamics simulations. The acceptor binding site forms an unusually large and shallow binding site. Binding of the acceptor to the enzyme is much faster and stronger if the donor is present. This is due to strong hydrogen bonding between O6 of the proximal N-acetylglucosamine and an oxygen atom of the ß-phosphate of GDP-fucose. Therefore, we propose an ordered Bi Bi mechanism for FUT8 where the donor molecule binds first. No specific amino acid is present that could act as base during catalysis. Our results indicate a donor-assisted mechanism, where an oxygen of the ß-phosphate deprotonates the acceptor. Knowledge of the mechanism of FUT8 is now being used for rational design of targeted inhibitors to address metastasis and prognosis of carcinomas.


Assuntos
Fucosiltransferases/metabolismo , Simulação de Dinâmica Molecular , Sítios de Ligação , Sequência de Carboidratos , Catálise , Fucosiltransferases/química , Humanos , Modelos Moleculares , Peptídeos/química , Peptídeos/genética
18.
Biochim Biophys Acta ; 1820(12): 1915-25, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22982178

RESUMO

BACKGROUND: Fucosylation is essential for various biological processes including tumorigenesis, inflammation, cell-cell recognition and host-pathogen interactions. Biosynthesis of fucosylated glycans is accomplished by fucosyltransferases. The enzymatic product of core α1,6-fucosyltransferase (FUT8) plays a major role in a plethora of pathological conditions, e.g. in prognosis of hepatocellular carcinoma and in colon cancer. Detailed knowledge of the binding mode of its substrates is required for the design of molecules that can modulate the activity of the enzyme. METHODS: We provide a detailed description of binding interactions of human FUT8 with its natural donor substrate GDP-fucose and related compounds. GDP-Fuc was placed in FUT8 by structural analogy to the structure of protein-O-fucosyltransferase (cePOFUT) co-crystallized with GDP-Fuc. The epitope of the donor substrate bound to FUT8 was determined by STD NMR. The in silico model is further supported by experimental data from SPR binding assays. The complex was optimized by molecular dynamics simulations. RESULTS: Guanine is specifically recognized by His363 and Asp453. Furthermore, the pyrophosphate is tightly bound via numerous hydrogen bonds and contributes affinity to a major part. Arg365 was found to bind both the ß-phosphate and the fucose moiety at the same time. CONCLUSIONS: Discovery of a novel structural analogy between cePOFUT and FUT8 allows the placement of the donor substrate GDP-Fuc. The positioning was confirmed by various experimental and computational techniques. GENERAL SIGNIFICANCE: The model illustrates details of the molecular basis of substrate recognition for a human fucosyltransferase for the first time and, thus, provides a basis for structure-based design of inhibitors.


Assuntos
Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/metabolismo , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Catálise , Fucosiltransferases/química , Fucosiltransferases/genética , Fucosiltransferases/isolamento & purificação , Humanos , Simulação de Dinâmica Molecular , Conformação Proteica , Ressonância de Plasmônio de Superfície
19.
J Biol Chem ; 287(20): 16709-19, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22451675

RESUMO

α-L-fucosyl residues attached at the non-reducing ends of glycoconjugates constitute histo-blood group antigens Lewis (Le) and ABO and play fundamental roles in various biological processes. Therefore, establishing a method for synthesizing the antigens is important for functional glycomics studies. However, regiospecific synthesis of glycosyl linkages, especially α-L-fucosyl linkages, is quite difficult to control both by chemists and enzymologists. Here, we generated an α-L-fucosynthase that specifically introduces Le(a) and Le(x) antigens into the type-1 and type-2 chains, respectively; i.e. the enzyme specifically accepts the disaccharide structures (Galß1-3/4GlcNAc) at the non-reducing ends and attaches a Fuc residue via an α-(1,4/3)-linkage to the GlcNAc. X-ray crystallographic studies revealed the structural basis of this strict regio- and acceptor specificity, which includes the induced fit movement of the catalytically important residues, and the difference between the active site structures of 1,3-1,4-α-L-fucosidase (EC 3.2.1.111) and α-L-fucosidase (EC 3.2.1.51) in glycoside hydrolase family 29. The glycosynthase developed in this study should serve as a potentially powerful tool to specifically introduce the Le(a/x) epitopes onto labile glycoconjugates including glycoproteins. Mining glycosidases with strict specificity may represent the most efficient route to the specific synthesis of glycosidic bonds.


Assuntos
Proteínas de Bactérias/química , Bifidobacterium/enzimologia , Fucose/química , Fucosiltransferases/química , Oligossacarídeos/química , Proteínas de Bactérias/genética , Bifidobacterium/genética , Domínio Catalítico , Epitopos/química , Fucosiltransferases/genética , Humanos , Antígenos do Grupo Sanguíneo de Lewis
20.
Protein Cell ; 2(1): 41-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21337008

RESUMO

Plants are known to be efficient hosts for the production of mammalian therapeutic proteins. However, plants produce complex N-glycans bearing ß1,2-xylose and core α1,3-fucose residues, which are absent in mammals. The immunogenicity and allergenicity of plant-specific Nglycans is a key concern in mammalian therapy. In this study, we amplified the sequences of 2 plant-specific glycosyltransferases from Nicotiana tabacum L. cv Bright Yellow 2 (BY2), which is a well-established cell line widely used for the expression of therapeutic proteins. The expression of the endogenous xylosyltranferase (XylT) and fucosyltransferase (FucT) was downregulated by using RNA interference (RNAi) strategy. The xylosylated and core fucosylated N-glycans were significantly, but not completely, reduced in the glycoengineered lines. However, these RNAi-treated cell lines were stable and viable and did not exhibit any obvious phenotype. Therefore, this study may provide an effective and promising strategy to produce recombinant glycoproteins in BY2 cells with humanized N-glycoforms to avoid potential immunogenicity.


Assuntos
Regulação para Baixo , Epitopos/genética , Epitopos/imunologia , Glicoproteínas/genética , Nicotiana/citologia , Nicotiana/genética , Engenharia de Proteínas/métodos , Sequência de Aminoácidos , Western Blotting , Sequência de Carboidratos , Linhagem Celular , Clonagem Molecular , DNA Complementar/genética , Fucose/metabolismo , Fucosiltransferases/química , Fucosiltransferases/deficiência , Fucosiltransferases/genética , Fucosiltransferases/imunologia , Glicoproteínas/química , Glicoproteínas/imunologia , Dados de Sequência Molecular , Pentosiltransferases/química , Pentosiltransferases/deficiência , Pentosiltransferases/genética , Pentosiltransferases/imunologia , Polissacarídeos/química , Polissacarídeos/imunologia , Interferência de RNA , Especificidade da Espécie , Xilose/metabolismo , UDP Xilose-Proteína Xilosiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA