Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(47): 12023-12027, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30397147

RESUMO

The N-glycans attached to the Fab and Fc domains play distinct roles in modulating the functions of antibodies. However, posttranslational site-selective modifications of glycans in antibodies and other multiply glycosylated proteins remain a challenging task. Here, we report a chemoenzymatic method that permits independent manipulation of the Fab and Fc N-glycans, using cetuximab as a model therapeutic monoclonal antibody. Taking advantage of the substrate specificity of three endoglycosidases (Endo-S, Endo-S2, and Endo-F3) and their glycosynthase mutants, together with an unexpected substrate site-selectivity of a bacterial α1,6-fucosidase from Lactobacillus casei (AlfC), we were able to synthesize an optimal homogeneous glycoform of cetuximab in which the heterogeneous and immunogenic Fab N-glycans were replaced with a single sialylated N-glycan, and the core-fucosylated Fc N-glycans were remodeled with a nonfucosylated and fully galactosylated N-glycan. The glycoengineered cetuximab demonstrated increased affinity for the FcγIIIa receptor and significantly enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) activity.


Assuntos
Fragmentos Fab das Imunoglobulinas/metabolismo , Fragmentos Fc das Imunoglobulinas/metabolismo , Engenharia de Proteínas/métodos , Anticorpos Monoclonais/química , Citotoxicidade Celular Dependente de Anticorpos/genética , Citotoxicidade Celular Dependente de Anticorpos/fisiologia , Cetuximab/metabolismo , Glicosídeo Hidrolases/metabolismo , Glicosilação , Humanos , Polissacarídeos/metabolismo , Especificidade por Substrato
2.
J Biol Chem ; 291(17): 9356-70, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-26966183

RESUMO

Chemoenzymatic synthesis is emerging as a promising approach to the synthesis of homogeneous glycopeptides and glycoproteins highly demanded for functional glycomics studies, but its generality relies on the availability of a range of enzymes with high catalytic efficiency and well defined substrate specificity. We describe in this paper the discovery of glycosynthase mutants derived from Elizabethkingia meningoseptica endoglycosidase F3 (Endo-F3) of the GH18 family, which are devoid of the inherent hydrolytic activity but are able to take glycan oxazolines for transglycosylation. Notably, the Endo-F3 D165A and D165Q mutants demonstrated high acceptorsubstrate specificity toward α1,6-fucosyl-GlcNAc-Asn or α1,6-fucosyl-GlcNAc-polypeptide in transglycosylation, enabling a highly convergent synthesis of core-fucosylated, complex CD52 glycopeptide antigen. The Endo-F3 mutants were able to use both bi- and triantennary glycan oxazolines as substrates for transglycosylation, in contrast to previously reported endoglycosidases derived from Endo-S, Endo-M, Endo-D, and Endo-A mutants that could not recognize triantennary N-glycans. Using rituximab as a model system, we have further demonstrated that the Endo-F3 mutants are highly efficient for glycosylation remodeling of monoclonal antibodies to produce homogeneous intact antibody glycoforms. Interestingly, the new triantennary glycan glycoform of antibody showed much higher affinity for galectin-3 than that of the commercial antibody. The Endo-F3 mutants represent the first endoglycosidase-based glycosynthases capable of transferring triantennary complex N-glycans, which would be very useful for glycoprotein synthesis and glycosylation remodeling of antibodies.


Assuntos
Proteínas de Bactérias , Flavobacteriaceae , Glicoproteínas , Glicosiltransferases , Anticorpos Antibacterianos/química , Anticorpos Monoclonais Murinos/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Flavobacteriaceae/genética , Flavobacteriaceae/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilação , Glicosiltransferases/genética , Glicosiltransferases/metabolismo
3.
Proc Natl Acad Sci U S A ; 111(18): 6714-9, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24753590

RESUMO

To evade host immune mechanisms, many bacteria secrete immunomodulatory enzymes. Streptococcus pyogenes, one of the most common human pathogens, secretes a large endoglycosidase, EndoS, which removes carbohydrates in a highly specific manner from IgG antibodies. This modification renders antibodies incapable of eliciting host effector functions through either complement or Fc γ receptors, providing the bacteria with a survival advantage. On account of this antibody-specific modifying activity, EndoS is being developed as a promising injectable therapeutic for autoimmune diseases that rely on autoantibodies. Additionally, EndoS is a key enzyme used in the chemoenzymatic synthesis of homogenously glycosylated antibodies with tailored Fc γ receptor-mediated effector functions. Despite the tremendous utility of this enzyme, the molecular basis of EndoS specificity for, and processing of, IgG antibodies has remained poorly understood. Here, we report the X-ray crystal structure of EndoS and provide a model of its encounter complex with its substrate, the IgG1 Fc domain. We show that EndoS is composed of five distinct protein domains, including glycosidase, leucine-rich repeat, hybrid Ig, carbohydrate binding module, and three-helix bundle domains, arranged in a distinctive V-shaped conformation. Our data suggest that the substrate enters the concave interior of the enzyme structure, is held in place by the carbohydrate binding module, and that concerted conformational changes in both enzyme and substrate are required for subsequent antibody deglycosylation. The EndoS structure presented here provides a framework from which novel endoglycosidases could be engineered for additional clinical and biotechnological applications.


Assuntos
Proteínas de Bactérias/química , Glicosídeo Hidrolases/química , Imunoglobulina G/metabolismo , Streptococcus pyogenes/enzimologia , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Glicosídeo Hidrolases/imunologia , Glicosídeo Hidrolases/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/química , Fatores Imunológicos/química , Fatores Imunológicos/metabolismo , Modelos Moleculares , Conformação Proteica , Estrutura Terciária de Proteína , Espalhamento a Baixo Ângulo , Streptococcus pyogenes/patogenicidade , Especificidade por Substrato , Difração de Raios X
4.
Bioorg Med Chem ; 21(8): 2262-2270, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23477942

RESUMO

A chemoenyzmatic method for direct glycosylation of polypeptides is described. The method consists of two site-specific enzymatic glycosylation steps: introduction of a glucose moiety at the consensus N-glycosylation sequence (NXS/T) in a polypeptide by an N-glycosyltransferase (NGT) and attachment of a complex N-glycan to the glucose primer by an endoglycosidase (ENGase)-catalyzed transglycosylation. Our experiments demonstrated that a relatively small excess of the UDP-Glc (the donor substrate) was sufficient for an effective glucosylation of polypeptides by the NGT, and different high-mannose and complex type N-glycans could be readily transferred to the glucose moiety by ENGases to provide full-size glycopeptides. The usefulness of the chemoenzymatic method was exemplified by an efficient synthesis of a complex glycoform of polypeptide C34, a potent HIV inhibitor derived from HIV-1 gp41. A comparative study indicated that the Glc-peptide was equally efficient as the natural GlcNAc-peptide to serve as an acceptor in the transglycosylation with sugar oxazoline as the donor substrate. Interestingly, the Glc-Asn linked glycopeptide was completely resistant to PNGase F digestion, in contrast to the GlcNAc-Asn linked natural glycopeptide that is an excellent substrate for hydrolysis. In addition, the Glc-Asn linked glycopeptide showed at least 10-fold lower hydrolytic activity toward Endo-M than the natural GlcNAc-Asn linked glycopeptide. The chemoenzymatic glycosylation method described here provides an efficient way to introducing complex N-glycans into polypeptides, for gain of novel properties that could be valuable for drug discovery.


Assuntos
Proteínas de Bactérias/química , Glucosiltransferases/química , Glicopeptídeos/síntese química , Peptídeos/química , Polissacarídeos/química , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Catálise , Glucosiltransferases/metabolismo , Glicopeptídeos/química , Glicosilação , Dados de Sequência Molecular , Peptídeos/metabolismo , Polissacarídeos/metabolismo
5.
Artigo em Inglês | MEDLINE | ID: mdl-24316841

RESUMO

Endoglycosidase S (EndoS) is an enzyme secreted by Streptococcus pyogenes that specifically hydrolyzes the ß-1,4-di-N-acetylchitobiose core glycan on immunoglobulin G (IgG) antibodies. One of the most common human pathogens and the cause of group A streptococcal infections, S. pyogenes secretes EndoS in order to evade the host immune system by rendering IgG effector mechanisms dysfunctional. On account of its specificity for IgG, EndoS has also been used extensively for chemoenzymatic synthesis of homogeneous IgG glycoprotein preparations and is being developed as a novel therapeutic for a wide range of autoimmune diseases. The structural basis of its enzymatic activity and substrate specificity, however, remains unknown. Here, the purification and crystallization of EndoS are reported. Using traditional hanging-drop and sitting-drop vapor-diffusion crystallization, crystals of EndoS were grown that diffracted to a maximum of 3.5 Å resolution but suffered from severe anisotropy, the data from which could only be reasonably processed to 7.5 Å resolution. When EndoS was crystallized by liquid-liquid diffusion, it was possible to grow crystals with a different space group to those obtained by vapor diffusion. Crystals of wild-type endoglycosidase and glycosynthase constructs of EndoS grown by liquid-liquid diffusion diffracted to 2.6 and 1.9 Å resolution, respectively, with a greatly diminished anisotropy. Despite extensive efforts, the failure to reproduce these liquid-liquid diffusion-grown crystals by vapor diffusion suggests that these crystallization methods each sample a distinct crystallization space.


Assuntos
Acetilglucosaminidase/química , Proteínas de Bactérias/química , Imunoglobulina G/química , Streptococcus pyogenes/química , Acetilglucosaminidase/genética , Acetilglucosaminidase/metabolismo , Anisotropia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cristalização , Cristalografia por Raios X , Difusão , Dissacarídeos/química , Dissacarídeos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Imunoglobulina G/metabolismo , Transição de Fase , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Especificidade por Substrato
6.
J Bacteriol ; 193(9): 2089-96, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21398546

RESUMO

The heparin-binding hemagglutinin adhesin (HBHA) is a surface adhesin on the human pathogen Mycobacterium tuberculosis. Previously, it has been shown that HBHA exists as a dimer in solution. We investigated the detailed nature of this dimer using circular dichroism spectroscopy and analytical ultracentrifugation techniques. We demonstrate that the heparan sulfate (HS) binding region does not play a role in dimerization in solution, while the linker region between the predicted N-terminal coiled-coil and the C-terminal HS binding region does affect dimer stability. The majority of contacts responsible for dimerization, folding, and stability lie within the predicted coiled-coil region of HBHA, while the N-terminal helix preceding the coiled-coil appears to trigger the folding and dimerization of HBHA. Constructs lacking this initial helix or containing site-specific mutations produce nonhelical monomers in solution. Thus, we show that HBHA dimerization and folding are linked and that the N-terminal region of this cell surface adhesin triggers the formation of an HBHA coiled-coil dimer.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteínas de Membrana/metabolismo , Mycobacterium tuberculosis/metabolismo , Dobramento de Proteína , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Clonagem Molecular , Proteínas de Membrana/genética , Dados de Sequência Molecular , Conformação Proteica
7.
J Mol Biol ; 426(18): 3166-3179, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25036289

RESUMO

Immunoglobulin G (IgG) is a central mediator of host defense due to its ability to recognize and eliminate pathogens. The recognition and effector responses are encoded on distinct regions of IgGs. The diversity of the antigen recognition Fab domains accounts for IgG's ability to bind with high specificity to essentially any antigen. Recent studies have indicated that the Fc effector domain also displays considerable heterogeneity, accounting for its complex effector functions of inflammation, modulation, and immune suppression. Therapeutic anti-tumor antibodies, for example, require the pro-inflammatory properties of the IgG Fc to eliminate tumor cells, while the anti-inflammatory activity of intravenous IgG requires specific Fc glycans for activity. In particular, the anti-inflammatory activity of intravenous IgG is ascribed to a small population of IgGs in which the Asn297-linked complex N-glycans attached to each Fc CH2 domain include terminal α2,6-linked sialic acids. We used chemoenzymatic glycoengineering to prepare fully disialylated IgG Fc and solved its crystal structure. Comparison of the structures of asialylated Fc, sialylated Fc, and F241A Fc, a mutant that displays increased glycan sialylation, suggests that increased conformational flexibility of the CH2 domain is associated with the switch from pro-inflammatory to anti-inflammatory activity of the Fc.


Assuntos
Anti-Inflamatórios/química , Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/química , Modelos Moleculares , Polissacarídeos/metabolismo , Anti-Inflamatórios/isolamento & purificação , Anti-Inflamatórios/metabolismo , Antígenos/imunologia , Linhagem Celular , Cristalografia por Raios X , Glicosilação , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/isolamento & purificação , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/genética , Imunoglobulina G/isolamento & purificação , Imunoglobulina G/metabolismo , Imunoglobulinas Intravenosas/química , Imunoglobulinas Intravenosas/genética , Imunoglobulinas Intravenosas/isolamento & purificação , Imunoglobulinas Intravenosas/metabolismo , Mutação , Estrutura Terciária de Proteína , Ácidos Siálicos/metabolismo
8.
ACS Chem Biol ; 7(1): 110-22, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22141574

RESUMO

Protein glycosylation is a common and complex posttranslational modification of proteins, which expands functional diversity while boosting structural heterogeneity. Glycoproteins, the end products of such a modification, are typically produced as mixtures of glycoforms possessing the same polypeptide backbone but differing in the site of glycosylation and/or in the structures of pendant glycans, from which single glycoforms are difficult to isolate. The urgent need for glycan-defined glycoproteins in both detailed structure-function relationship studies and therapeutic applications has stimulated an extensive interest in developing various methods for manipulating protein glycosylation. This review highlights emerging technologies that hold great promise in making a variety of glycan-defined glycoproteins, with a particular emphasis in the following three areas: specific glycoengineering of host biosynthetic pathways, in vitro chemoenzymatic glycosylation remodeling, and chemoselective and site-specific glycosylation of proteins.


Assuntos
Glicoproteínas/metabolismo , Polissacarídeos/química , Engenharia de Proteínas/métodos , Processamento de Proteína Pós-Traducional , Animais , Células CHO , Configuração de Carboidratos , Sequência de Carboidratos , Cricetinae , Escherichia coli , Expressão Gênica , Glicoconjugados/síntese química , Glicoconjugados/metabolismo , Glicoproteínas/genética , Glicosilação , Células HEK293 , Humanos , Isomerismo , Modelos Moleculares , Dados de Sequência Molecular , Células Vegetais , Polissacarídeos/metabolismo , Estrutura Secundária de Proteína , Relação Estrutura-Atividade , Leveduras
9.
Bioorg Med Chem Lett ; 16(14): 3802-5, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16678412

RESUMO

2,3-Dihydroxybenzohydroxamoyl adenylate (I) was prepared as a potential product analog inhibitor of EntE (EC# 2.7.7.58), a 2,3-dihydroxybenzoate AMP ligase from Escherichia coli that is required for the biosynthesis of enterobactin. This compound, obtained by the aqueous reaction of imidazole-activated adenosine 5'-phosphate and 2,3-dihydroxybenzohydroxamic acid, is a competitive inhibitor with a Ki value of 4.5 x 10(-9)M. Deletion of the catecholic 3-OH group of (I), in compound (II), reduced inhibitory activity by a factor of 3.5, whereas, removal of both the 3-OH and 2-OH groups, in (III), reduced inhibitory activity by a factor of approximately 2000. Acetohydroxamoyl adenylate (IV), in which the entire catechol moiety of (I) is replaced by a hydrogen atom, gave

Assuntos
Monofosfato de Adenosina/análogos & derivados , Enterobactina/biossíntese , Inibidores Enzimáticos/síntese química , Proteínas de Escherichia coli/antagonistas & inibidores , Hidroxibenzoatos/síntese química , Ligases/antagonistas & inibidores , Monofosfato de Adenosina/síntese química , Monofosfato de Adenosina/química , Monofosfato de Adenosina/metabolismo , Monofosfato de Adenosina/farmacologia , Ligação Competitiva , Catecóis/química , Inibidores Enzimáticos/farmacologia , Escherichia coli/enzimologia , Hidrogênio/química , Ácidos Hidroxâmicos/química , Hidroxibenzoatos/química , Hidroxibenzoatos/farmacologia , Imidazóis/química , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA