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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 298(1): 101454, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34838817

RESUMO

Glycoproteins are difficult to crystallize because they have heterogeneous glycans composed of multiple monosaccharides with considerable rotational freedom about their O-glycosidic linkages. Crystallographers studying N-glycoproteins often circumvent this problem by using ß1,2-N-acetylglucosaminyltransferase I (MGAT1)-deficient mammalian cell lines, which produce recombinant glycoproteins with immature N-glycans. These glycans support protein folding and quality control but can be removed using endo-ß-N-acetylglucosaminidase H (Endo H). Many crystallographers also use the baculovirus-insect cell system (BICS) to produce recombinant proteins for their work but have no access to an MGAT1-deficient insect cell line to facilitate glycoprotein crystallization in this system. Thus, we used BICS-specific CRISPR-Cas9 vectors to edit the Mgat1 gene of a rhabdovirus-negative Spodoptera frugiperda cell line (Sf-RVN) and isolated a subclone with multiple Mgat1 deletions, which we named Sf-RVNLec1. We found that Sf-RVN and Sf-RVNLec1 cells had identical growth properties and served equally well as hosts for baculovirus-mediated recombinant glycoprotein production. N-glycan profiling showed that a total endogenous glycoprotein fraction isolated from Sf-RVNLec1 cells had only immature and high mannose-type N-glycans. Finally, N-glycan profiling and endoglycosidase analyses showed that the vast majority of the N-glycans on three recombinant glycoproteins produced by Sf-RVNLec1 cells were Endo H-cleavable Man5GlcNAc2 structures. Thus, this study yielded a new insect cell line for the BICS that can be used to produce recombinant glycoproteins with Endo H-cleavable N-glycans. This will enable researchers to combine the high productivity of the BICS with the ability to deglycosylate recombinant glycoproteins, which will facilitate efforts to determine glycoprotein structures by X-ray crystallography.


Assuntos
Baculoviridae , Linhagem Celular , Insetos , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Sistemas CRISPR-Cas , Glicoproteínas/biossíntese , Glicoproteínas/genética , Humanos , Insetos/citologia , Insetos/genética , Insetos/metabolismo , Polissacarídeos/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
2.
Br J Cancer ; 123(11): 1633-1643, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32921792

RESUMO

BACKGROUND: Annexin A1 is expressed specifically on the tumour vasculature surface. Intravenously injected IF7 targets tumour vasculature via annexin A1. We tested the hypothesis that IF7 overcomes the blood-brain barrier and that the intravenously injected IF7C(RR)-SN38 eradicates brain tumours in the mouse. METHODS: (1) A dual-tumour model was generated by inoculating luciferase-expressing melanoma B16 cell line, B16-Luc, into the brain and under the skin of syngeneic C57BL/6 mice. IF7C(RR)-SN38 was injected intravenously daily at 7.0 µmoles/kg and growth of tumours was assessed by chemiluminescence using an IVIS imager. A similar dual-tumour model was generated with the C6-Luc line in immunocompromised SCID mice. (2) IF7C(RR)-SN38 formulated with 10% Solutol HS15 was injected intravenously daily at 2.5 µmoles/kg into two brain tumour mouse models: B16-Luc cells in C57BL/6 mice, and C6-Luc cells in nude mice. RESULTS: (1) Daily IF7C(RR)-SN38 injection suppressed tumour growth regardless of cell lines or mouse strains. (2) Daily injection of Solutol-formulated IF7C(RR)-SN38 led into complete disappearance of B16-Luc brain tumour in C57BL/6 mice, whereas this did not occur in C6-Luc in nude mice. CONCLUSIONS: IF7C(RR)-SN38 crosses the blood-brain barrier and suppresses growth of brain tumours in mouse models. Solutol HS15-formulated IF7C(RR)-SN38 may have promoted an antitumour immune response.


Assuntos
Anexina A1/metabolismo , Antineoplásicos/farmacologia , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas , Portadores de Fármacos/farmacologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Peptídeos , Ratos
3.
Nat Chem Biol ; 14(2): 156-162, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29251719

RESUMO

Vertebrate glycoproteins and glycolipids are synthesized in complex biosynthetic pathways localized predominantly within membrane compartments of the secretory pathway. The enzymes that catalyze these reactions are exquisitely specific, yet few have been extensively characterized because of challenges associated with their recombinant expression as functional products. We used a modular approach to create an expression vector library encoding all known human glycosyltransferases, glycoside hydrolases, and sulfotransferases, as well as other glycan-modifying enzymes. We then expressed the enzymes as secreted catalytic domain fusion proteins in mammalian and insect cell hosts, purified and characterized a subset of the enzymes, and determined the structure of one enzyme, the sialyltransferase ST6GalNAcII. Many enzymes were produced at high yields and at similar levels in both hosts, but individual protein expression levels varied widely. This expression vector library will be a transformative resource for recombinant enzyme production, broadly enabling structure-function studies and expanding applications of these enzymes in glycochemistry and glycobiology.


Assuntos
Perfilação da Expressão Gênica , Sialiltransferases/química , Animais , Baculoviridae/metabolismo , Cristalografia por Raios X , Monofosfato de Citidina/química , Vetores Genéticos , Glicosídeo Hidrolases/química , Glicosilação , Células HEK293 , Humanos , Insetos , Cinética , Proteínas Recombinantes/química , Sulfotransferases/química
4.
Biotechnol Bioeng ; 117(11): 3248-3264, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32662870

RESUMO

Cell lines derived from Trichoplusia ni (Tn) are widely used as hosts in the baculovirus-insect cell system (BICS). One advantage of Tn cell lines is they can produce recombinant proteins at higher levels than cell lines derived from other insects. However, Tn cell lines are persistently infected with an alphanodavirus, Tn5 cell-line virus (TnCLV), which reduces their utility as a host for the BICS. Several groups have isolated TnCLV-negative Tn cell lines, but none were thoroughly characterized and shown to be free of other adventitious viruses. Thus, we isolated and extensively characterized a new TnCLV-negative line, Tn-nodavirus-negative (Tn-NVN). Tn-NVN cells have no detectable TnCLV, no other previously identified viral contaminants of lepidopteran insect cell lines, and no sequences associated with any replicating virus or other viral adventitious agents. Tn-NVN cells tested negative for >60 species of Mycoplasma, Acholeplasma, Spiroplasma, and Ureaplasma. Finally, Tn-NVN cells grow well as a single-cell suspension culture in serum-free medium, produce recombinant proteins at levels similar to High Five™ cells, and do not produce recombinant glycoproteins with immunogenic core α1,3-fucosylation. Thus, Tn-NVN is a new, well-characterized TnCLV-negative cell line with several other features enhancing its utility as a host for the BICS.


Assuntos
Baculoviridae/genética , Técnicas de Cultura de Células/métodos , Lepidópteros , Animais , Linhagem Celular , Lepidópteros/citologia , Lepidópteros/genética , Lepidópteros/metabolismo , Lepidópteros/virologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
Proc Natl Acad Sci U S A ; 114(34): 9068-9073, 2017 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-28784806

RESUMO

The baculovirus-insect cell system (BICS) has been widely used to produce many different recombinant proteins for basic research and is being used to produce several biologics approved for use in human or veterinary medicine. Early BICS were technically complex and constrained by the relatively primordial nature of insect cell protein glycosylation pathways. Since then, recombination has been used to modify baculovirus vectors-which has simplified the system-and transform insect cells, which has enhanced its protein glycosylation capabilities. Now, CRISPR-Cas9 tools for site-specific genome editing are needed to facilitate further improvements in the BICS. Thus, in this study, we used various insect U6 promoters to construct CRISPR-Cas9 vectors and assessed their utility for site-specific genome editing in two insect cell lines commonly used as hosts in the BICS. We demonstrate the use of CRISPR-Cas9 to edit an endogenous insect cell gene and alter protein glycosylation in the BICS.


Assuntos
Baculoviridae/genética , Sistemas CRISPR-Cas , Edição de Genes/métodos , Insetos/genética , Animais , Sequência de Bases , Linhagem Celular , Glicosilação , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Insetos/citologia , Modelos Genéticos , Regiões Promotoras Genéticas/genética , RNA Nuclear Pequeno/genética , Homologia de Sequência do Ácido Nucleico , Células Sf9 , Spodoptera
6.
Protein Expr Purif ; 144: 25-32, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29133148

RESUMO

Insect cells are widely used for recombinant protein expression, typically as hosts for recombinant baculovirus vectors, but also for plasmid-mediated transient transfection or stable genetic transformation. Insect cells are used to express proteins for research, as well as to manufacture biologicals for human and veterinary medicine. Recently, several insect cell lines used for recombinant protein expression were found to be persistently infected with adventitious viruses. This has raised questions about how these infections might affect research performed using those cell lines. Furthermore, these findings raised serious concerns about the safety of biologicals produced using those cell lines. In response, new insect cell lines lacking adventitious viruses have been isolated for use as improved research tools and safer biological manufacturing platforms. Here, we review the scientific and patent literature on adventitious viruses found in insect cell lines, affected cell lines, and new virus-free cell lines.


Assuntos
Clonagem Molecular/métodos , Vetores Genéticos , Insetos/virologia , Proteínas Recombinantes/genética , Vírus/genética , Animais , Linhagem Celular , Expressão Gênica
7.
Glycobiology ; 26(4): 360-76, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26610890

RESUMO

A large family of UDP-GalNAc:polypeptide GalNAc transferases (ppGalNAc-Ts) initiates and defines sites of mucin-type Ser/Thr-O-GalNAc glycosylation. Family members have been classified into peptide- and glycopeptide-preferring subfamilies, although both families possess variable activities against glycopeptide substrates. All but one isoform contains a C-terminal carbohydrate-binding lectin domain whose roles in modulating glycopeptide specificity is just being understood. We have previously shown for several peptide-preferring isoforms that the presence of a remote Thr-O-GalNAc, 6-17 residues from a Ser/Thr acceptor site, may enhance overall catalytic activity in an N- or C-terminal direction. This enhancement varies with isoform and is attributed to Thr-O-GalNAc interactions at the lectin domain. We now report on the glycopeptide substrate utilization of a series of glycopeptide (human-ppGalNAc-T4, T7, T10, T12 and fly PGANT7) and peptide-preferring transferases (T2, T3 and T5) by exploiting a series of random glycopeptide substrates designed to probe the functions of their catalytic and lectin domains. Glycosylation was observed at the -3, -1 and +1 residues relative to a neighboring Thr-O-GalNAc, depending on isoform, which we attribute to specific Thr-O-GalNAc binding at the catalytic domain. Additionally, these glycopeptide-preferring isoforms show remote lectin domain-assisted Thr-O-GalNAc enhancements that vary from modest to none. We conclude that the glycopeptide specificity of the glycopeptide-preferring isoforms predominantly resides in their catalytic domain but may be further modulated by remote lectin domain interactions. These studies further demonstrate that both domains of the ppGalNAc-Ts have specialized and unique functions that work in concert to control and order mucin-type O-glycosylation.


Assuntos
Glicopeptídeos/química , Lectinas/química , Mucinas/química , Sialiltransferases/química , Sequência de Aminoácidos/genética , Sítios de Ligação , Carboidratos/química , Carboidratos/genética , Domínio Catalítico , Fucose/análogos & derivados , Fucose/química , Glicopeptídeos/biossíntese , Glicopeptídeos/genética , Glicosilação , Humanos , Lectinas/genética , Mucinas/biossíntese , Mucinas/genética , Filogenia , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Sialiltransferases/genética , Especificidade por Substrato
8.
Protein Expr Purif ; 122: 45-55, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26923062

RESUMO

Cell lines derived from the fall armyworm, Spodoptera frugiperda (Sf), are widely used as hosts for recombinant protein production in the baculovirus-insect cell system (BICS). However, it was recently discovered that these cell lines are contaminated with a virus, now known as Sf-rhabdovirus [1]. The detection of this adventitious agent raised a potential safety issue that could adversely impact the BICS as a commercial recombinant protein production platform. Thus, we examined the properties of Sf-RVN, an Sf-rhabdovirus-negative Sf cell line, as a potential alternative host. Nested RT-PCR assays showed Sf-RVN cells had no detectable Sf-rhabdovirus over the course of 60 passages in continuous culture. The general properties of Sf-RVN cells, including their average growth rates, diameters, morphologies, and viabilities after baculovirus infection, were virtually identical to those of Sf9 cells. Baculovirus-infected Sf-RVN and Sf9 cells produced equivalent levels of three recombinant proteins, including an intracellular prokaryotic protein and two secreted eukaryotic glycoproteins, and provided similar N-glycosylation patterns. In fact, except for the absence of Sf-rhabdovirus, the only difference between Sf-RVN and Sf9 cells was SF-RVN produced higher levels of infectious baculovirus progeny. These results show Sf-RVN cells can be used as improved, alternative hosts to circumvent the potential safety hazard associated with the use of Sf-rhabdovirus-contaminated Sf cells for recombinant protein manufacturing with the BICS.


Assuntos
Baculoviridae/genética , Clonagem Molecular , Mycoplasma/isolamento & purificação , Rhabdoviridae/isolamento & purificação , Spodoptera/citologia , Spodoptera/virologia , Animais , Clonagem Molecular/métodos , Eritropoetina/genética , Expressão Gênica , Humanos , Proteínas Recombinantes/genética , Células Sf9 , Spodoptera/genética
9.
Biologicals ; 44(4): 219-225, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27236849

RESUMO

Spodoptera frugiperda (Sf) cell lines are used to produce several biologicals for human and veterinary use. Recently, it was discovered that all tested Sf cell lines are persistently infected with Sf-rhabdovirus, a novel rhabdovirus. As part of an effort to search for other adventitious viruses, we searched the Sf cell genome and transcriptome for sequences related to Sf-rhabdovirus. To our surprise, we found intact Sf-rhabdovirus N- and P-like ORFs, and partial Sf-rhabdovirus G- and L-like ORFs. The transcribed and genomic sequences matched, indicating the transcripts were derived from the genomic sequences. These appear to be endogenous viral elements (EVEs), which result from the integration of partial viral genetic material into the host cell genome. It is theoretically impossible for the Sf-rhabdovirus-like EVEs to produce infectious virus particles as 1) they are disseminated across 4 genomic loci, 2) the G and L ORFs are incomplete, and 3) the M ORF is missing. Our finding of transcribed virus-like sequences in Sf cells underscores that MPS-based searches for adventitious viruses in cell substrates used to manufacture biologics should take into account both genomic and transcribed sequences to facilitate the identification of transcribed EVE's, and to avoid false positive detection of replication-competent adventitious viruses.


Assuntos
Genes Virais/genética , Genoma de Inseto/genética , Spodoptera/genética , Transcrição Gênica , Transcriptoma/genética , Animais , Linhagem Celular , Retrovirus Endógenos/genética , Regulação Viral da Expressão Gênica , Fases de Leitura Aberta/genética , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rhabdoviridae/genética , Spodoptera/citologia , Proteínas Virais/classificação , Proteínas Virais/genética , Vírion/genética , Vírus/genética
10.
J Bacteriol ; 197(24): 3760-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26391208

RESUMO

UNLABELLED: The sialyl-T antigen sialylα2-3Galß1-3GalNAc is a common O-glycan structure in human glycoproteins and is synthesized by sialyltransferase ST3Gal1. The enterohemorrhagic Escherichia coli serotype O104 has the rare ability to synthesize a sialyl-T antigen mimic. We showed here that the wbwA gene of the E. coli O104 antigen synthesis gene cluster encodes an α2,3-sialyltransferase WbwA that transfers sialic acid from CMP-sialic acid to Galß1-3GalNAcα-diphosphate-lipid acceptor. Nuclear magnetic resonance (NMR) analysis of purified WbwA enzyme reaction product indicated that the sialyl-T antigen sialylα2-3Galß1-3GalNAcα-diphosphate-lipid was synthesized. We showed that the conserved His-Pro (HP) motif and Glu/Asp residues of two EDG motifs in WbwA are important for the activity. The characterization studies showed that WbwA from E. coli O104 is a monofunctional α2,3-sialyltransferase and is distinct from human ST3Gal1 as well as all other known sialyltransferases due to its unique acceptor specificity. This work contributes to knowledge of the biosynthesis of bacterial virulence factors. IMPORTANCE: This is the first characterization of a sialyltransferase involved in the synthesis of an O antigen in E. coli. The enzyme contributes to the mimicry of human sialyl-T antigen and has unique substrate specificity but very little sequence identity to other sialyltransferases. Thus, the bacterial sialyltransferase is related to the human counterpart only by the similarity of biochemical activity.


Assuntos
Escherichia coli Êntero-Hemorrágica/metabolismo , Proteínas de Escherichia coli/química , Antígenos O/biossíntese , Sialiltransferases/química , Sialiltransferases/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Escherichia coli Êntero-Hemorrágica/genética , Proteínas de Escherichia coli/genética , Humanos , Ácido N-Acetilneuramínico/química , Ressonância Magnética Nuclear Biomolecular , Análise de Sequência de DNA , Sialiltransferases/genética , beta-Galactosídeo alfa-2,3-Sialiltransferase
11.
J Virol ; 88(3): 1771-80, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24257615

RESUMO

Alphaviruses are mosquito-borne viruses that cause significant disease in animals and humans. Western equine encephalitis virus (WEEV) and eastern equine encephalitis virus (EEEV), two New World alphaviruses, can cause fatal encephalitis, and EEEV is a select agent of concern in biodefense. However, we have no antiviral therapies against alphaviral disease, and current vaccine strategies target only a single alphavirus species. In an effort to develop new tools for a broader response to outbreaks, we designed and tested a novel alphavirus vaccine comprised of cationic lipid nucleic acid complexes (CLNCs) and the ectodomain of WEEV E1 protein (E1ecto). Interestingly, we found that the CLNC component, alone, had therapeutic efficacy, as it increased survival of CD-1 mice following lethal WEEV infection. Immunization with the CLNC-WEEV E1ecto mixture (lipid-antigen-nucleic acid complexes [LANACs]) using a prime-boost regimen provided 100% protection in mice challenged with WEEV subcutaneously, intranasally, or via mosquito. Mice immunized with LANACs mounted a strong humoral immune response but did not produce neutralizing antibodies. Passive transfer of serum from LANAC E1ecto-immunized mice to nonimmune CD-1 mice conferred protection against WEEV challenge, indicating that antibody is sufficient for protection. In addition, the LANAC E1ecto immunization protocol significantly increased survival of mice following intranasal or subcutaneous challenge with EEEV. In summary, our LANAC formulation has therapeutic potential and is an effective vaccine strategy that offers protection against two distinct species of alphavirus irrespective of the route of infection. We discuss plausible mechanisms as well the potential utility of our LANAC formulation as a pan-alphavirus vaccine.


Assuntos
Antígenos Virais/imunologia , Vírus da Encefalite Equina do Leste/fisiologia , Vírus da Encefalite Equina do Oeste/fisiologia , Encefalomielite Equina/prevenção & controle , Lipossomos/imunologia , Ácidos Nucleicos/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/química , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/administração & dosagem , Antígenos Virais/química , Antígenos Virais/genética , Vírus da Encefalite Equina do Leste/imunologia , Vírus da Encefalite Equina do Oeste/imunologia , Encefalomielite Equina/imunologia , Encefalomielite Equina/virologia , Feminino , Humanos , Imunização , Lipossomos/administração & dosagem , Lipossomos/química , Camundongos , Ácidos Nucleicos/administração & dosagem , Ácidos Nucleicos/química , Proteínas Virais/administração & dosagem , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/química , Vacinas Virais/genética
12.
Proc Natl Acad Sci U S A ; 109(3): 923-8, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22215590

RESUMO

The development of a spider silk-manufacturing process is of great interest. However, there are serious problems with natural manufacturing through spider farming, and standard recombinant protein production platforms have provided limited progress due to their inability to assemble spider silk proteins into fibers. Thus, we used piggyBac vectors to create transgenic silkworms encoding chimeric silkworm/spider silk proteins. The silk fibers produced by these animals were composite materials that included chimeric silkworm/spider silk proteins integrated in an extremely stable manner. Furthermore, these composite fibers were, on average, tougher than the parental silkworm silk fibers and as tough as native dragline spider silk fibers. These results demonstrate that silkworms can be engineered to manufacture composite silk fibers containing stably integrated spider silk protein sequences, which significantly improve the overall mechanical properties of the parental silkworm silk fibers.


Assuntos
Bombyx/genética , Genes de Insetos/genética , Fenômenos Mecânicos , Seda/genética , Aranhas/genética , Animais , Animais Geneticamente Modificados , Elementos de DNA Transponíveis/genética , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/metabolismo
13.
Proteomics ; 14(1): 87-92, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24174266

RESUMO

The relative amount of high mannose structures within an N-glycomic pool differs from one source to another, but quite often it predominates over the larger size complex type structures carrying biologically important glyco-epitopes. An efficient method to separate these two classes of N-glycans would significantly aid in detecting the lower abundant components by MS. Capitalizing on an initial observation that only high mannose type structures were recovered in the flow-through fraction when peptide-N-glycosidase F digested peptides were passed through a C18 cartridge in 0.1% formic acid, we demonstrated here that native complex type N-glycans can be retained by C18 cartridge and to be efficiently separated from both the smaller high mannose type structures, as well as de-N-glycosylated peptides by stepwise elution with increasing ACN concentration. The weak retention of the largely hydrophilic N-glycans on C18 resin is dependent not only on size but also increased by the presence of α6-fucosylation. This was shown by comparing the resulting N-glycomic profiles of the washed and low-ACN eluted fractions derived from both a human cancer cell line and an insect cell line.


Assuntos
Glicômica/métodos , Glicopeptídeos/análise , Glicopeptídeos/química , Manose/isolamento & purificação , Sequência de Carboidratos , Manose/química , Espectrometria de Massas
14.
J Biol Chem ; 288(27): 19900-14, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23689369

RESUMO

Mucin type O-glycosylation is initiated by a large family of polypeptide GalNAc transferases (ppGalNAc Ts) that add α-GalNAc to the Ser and Thr residues of peptides. Of the 20 human isoforms, all but one are composed of two globular domains linked by a short flexible linker: a catalytic domain and a ricin-like lectin carbohydrate binding domain. Presently, the roles of the catalytic and lectin domains in peptide and glycopeptide recognition and specificity remain unclear. To systematically study the role of the lectin domain in ppGalNAc T glycopeptide substrate utilization, we have developed a series of novel random glycopeptide substrates containing a single GalNAc-O-Thr residue placed near either the N or C terminus of the glycopeptide substrate. Our results reveal that the presence and N- or C-terminal placement of the GalNAc-O-Thr can be important determinants of overall catalytic activity and specificity that differ between transferase isoforms. For example, ppGalNAc T1, T2, and T14 prefer C-terminally placed GalNAc-O-Thr, whereas ppGalNAc T3 and T6 prefer N-terminally placed GalNAc-O-Thr. Several transferase isoforms, ppGalNAc T5, T13, and T16, display equally enhanced N- or C-terminal activities relative to the nonglycosylated control peptides. This N- and/or C-terminal selectivity is presumably due to weak glycopeptide binding to the lectin domain, whose orientation relative to the catalytic domain is dynamic and isoform-dependent. Such N- or C-terminal glycopeptide selectivity provides an additional level of control or fidelity for the O-glycosylation of biologically significant sites and suggests that O-glycosylation may in some instances be exquisitely controlled.


Assuntos
Glicopeptídeos , Lectinas , N-Acetilgalactosaminiltransferases , Catálise , Glicopeptídeos/química , Glicopeptídeos/genética , Glicopeptídeos/metabolismo , Glicosilação , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , N-Acetilgalactosaminiltransferases/química , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Estrutura Terciária de Proteína , Especificidade por Substrato/fisiologia , Polipeptídeo N-Acetilgalactosaminiltransferase
15.
Biochim Biophys Acta ; 1830(8): 4274-81, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23578692

RESUMO

BACKGROUND: Modifications of proteins by O-glycosylation determine many of the properties and functions of proteins. We wish to understand the mechanisms of O-glycosylation and develop inhibitors that could affect glycoprotein functions and alter cellular behavior. METHODS: We expressed recombinant soluble human Gal- and GlcNAc-transferases that synthesize the O-glycan cores 1 to 4 and are critical for the overall structures of O-glycans. We determined the properties and substrate specificities of these enzymes using synthetic acceptor substrate analogs. Compounds that were inactive as substrates were tested as inhibitors. RESULTS: Enzymes significantly differed in their recognition of the sugar moieties and aglycone groups of substrates. Core 1 synthase was active with glycopeptide substrates but GlcNAc-transferases preferred substrates with hydrophobic aglycone groups. Chemical modifications of the acceptors shed light on enzyme-substrate interactions. Core 1 synthase was weakly inhibited by its substrate analog benzyl 2-butanamido-2-deoxy-α-d-galactoside while two of the three GlcNAc-transferases were selectively and potently inhibited by bis-imidazolium salts which are not substrate analogs. CONCLUSIONS: This work delineates the distinct specificities and properties of the enzymes that synthesize the common O-glycan core structures 1 to 4. New inhibitors were found that could selectively inhibit the synthesis of cores 1, 2 and 3 but not core 4. GENERAL SIGNIFICANCE: These studies help our understanding of the mechanisms of action of enzymes critical for O-glycosylation. The results may be useful for the re-engineering of O-glycosylation to determine the roles of O-glycans and the enzymes critical for O-glycosylation, and for biotechnology with potential therapeutic applications.


Assuntos
Galactosiltransferases/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Polissacarídeos/biossíntese , Galactosiltransferases/antagonistas & inibidores , Galactosiltransferases/química , Glicosilação , Humanos , N-Acetilglucosaminiltransferases/antagonistas & inibidores , N-Acetilglucosaminiltransferases/química , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
16.
Glycobiology ; 24(3): 325-40, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24362443

RESUMO

Glycosylation is an important attribute of baculovirus-insect cell expression systems, but some insect cell lines produce core α1,3-fucosylated N-glycans, which are highly immunogenic and render recombinant glycoproteins unsuitable for human use. To address this problem, we exploited a bacterial enzyme, guanosine-5'-diphospho (GDP)-4-dehydro-6-deoxy-d-mannose reductase (Rmd), which consumes the GDP-l-fucose precursor. We expected this enzyme to block glycoprotein fucosylation by blocking the production of GDP-l-fucose, the donor substrate required for this process. Initially, we engineered two different insect cell lines to constitutively express Rmd and isolated subclones with fucosylation-negative phenotypes. However, we found the fucosylation-negative phenotypes induced by Rmd expression were unstable, indicating that this host cell engineering approach is ineffective in insect systems. Thus, we constructed a baculovirus vector designed to express Rmd immediately after infection and facilitate the insertion of genes encoding any glycoprotein of interest for expression later after infection. We used this vector to produce a daughter encoding rituximab and found, in contrast to an Rmd-negative control, that insect cells infected with this virus produced a nonfucosylated form of this therapeutic antibody. These results indicate that our Rmd(+) baculoviral vector can be used to solve the immunogenic core α1,3-fucosylation problem associated with the baculovirus-insect cell system. In conjunction with existing glycoengineered insect cell lines, this vector extends the utility of the baculovirus-insect cell system to include therapeutic glycoprotein production. This new vector also extends the utility of the baculovirus-insect cell system to include the production of recombinant antibodies with enhanced effector functions, due to its ability to block core α1,6-fucosylation.


Assuntos
Proteínas de Bactérias/metabolismo , Vetores Genéticos/genética , Glicoproteínas/metabolismo , Cetona Oxirredutases/metabolismo , Animais , Proteínas de Bactérias/genética , Baculoviridae/genética , Biotecnologia/métodos , Fucose/metabolismo , Glicoproteínas/genética , Cetona Oxirredutases/genética , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Sf9 , Spodoptera
18.
J Proteome Res ; 12(8): 3707-20, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23848607

RESUMO

Hemagglutinin (HA) is the major antigen in influenza vaccines, and glycosylation is known to influence its antigenicity. Embryonated hen eggs are traditionally used for influenza vaccine production, but vaccines produced in mammalian and insect cells were recently licensed. This raises the concern that vaccines produced with different cell systems might not be equivalent due to differences in their glycosylation patterns. Thus, we developed an analytical method to monitor vaccine glycosylation through a combination of nanoLC/MS(E) and quantitative MALDI-TOF MS permethylation profiling. We then used this method to examine glycosylation of HAs from two different influenza H5N1 strains produced in five different platforms, including hen eggs, three different insect cell lines (High Five, expresSF+ and glycoengineered expresSF+), and a human cell line (HEK293). Our results demonstrated that (1) sequon utilization is not necessarily equivalent in different cell types, (2) there are quantitative and qualitative differences in the overall N-glycosylation patterns and structures produced by different cell types, (3) ∼20% of the N-glycans on the HAs produced by High Five cells are core α1,3-fucosylated structures, which may be allergenic in humans, and (4) our method can be used to monitor differences in glycosylation during the cellular glycoengineering stages of vaccine development.


Assuntos
Glicômica , Hemaglutininas Virais/química , Vírus da Influenza A Subtipo H1N1/química , Virus da Influenza A Subtipo H5N1/química , Polissacarídeos/análise , Sequência de Aminoácidos , Animais , Sequência de Carboidratos , Embrião de Galinha , Galinhas , Glicosilação , Células HEK293 , Hemaglutininas Virais/metabolismo , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/biossíntese , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Dados de Sequência Molecular , Polissacarídeos/química , Células Sf9 , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Spodoptera , Zigoto/virologia
19.
J Biol Chem ; 287(10): 7084-97, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22238347

RESUMO

Man(α1-6)[GlcNAc(ß1-2)Man(α1-3)]ManGlcNAc(2) is a key branch point intermediate in the insect N-glycosylation pathway because it can be either trimmed by a processing ß-N-acetylglucosaminidase (FDL) to produce paucimannosidic N-glycans or elongated by N-acetylglucosaminyltransferase II (GNT-II) to produce complex N-glycans. N-acetylglucosaminyltransferase I (GNT-I) contributes to branch point intermediate production and can potentially reverse the FDL trimming reaction. However, there has been no concerted effort to evaluate the relationships among these three enzymes in any single insect system. Hence, we extended our previous studies on Spodoptera frugiperda (Sf) FDL to include GNT-I and -II. Sf-GNT-I and -II cDNAs were isolated, the predicted protein sequences were analyzed, and both gene products were expressed and their acceptor substrate specificities and intracellular localizations were determined. Sf-GNT-I transferred N-acetylglucosamine to Man(5)GlcNAc(2), Man(3)GlcNAc(2), and GlcNAc(ß1-2)Man(α1-6)[Man(α1-3)]ManGlcNAc(2), demonstrating its role in branch point intermediate production and its ability to reverse FDL trimming. Sf-GNT-II only transferred N-acetylglucosamine to Man(α1-6)[GlcNAc(ß1-2)Man(α1-3)]ManGlcNAc(2), demonstrating that it initiates complex N-glycan production, but cannot use Man(3)GlcNAc(2) to produce hybrid or complex structures. Fluorescently tagged Sf-GNT-I and -II co-localized with an endogenous Sf Golgi marker and Sf-FDL co-localized with Sf-GNT-I and -II, indicating that all three enzymes are Golgi resident proteins. Unexpectedly, fluorescently tagged Drosophila melanogaster FDL also co-localized with Sf-GNT-I and an endogenous Drosophila Golgi marker, indicating that it is a Golgi resident enzyme in insect cells. Thus, the substrate specificities and physical juxtapositioning of GNT-I, GNT-II, and FDL support the idea that these enzymes function at the N-glycan processing branch point and are major factors determining the net outcome of the insect cell N-glycosylation pathway.


Assuntos
Complexo de Golgi/enzimologia , Proteínas de Insetos/metabolismo , Insetos/enzimologia , N-Acetilglucosaminiltransferases/metabolismo , Oligossacarídeos/biossíntese , Acetilglucosamina/genética , Acetilglucosamina/metabolismo , Animais , Sequência de Bases , DNA Complementar/genética , Glicosilação , Complexo de Golgi/genética , Proteínas de Insetos/genética , Insetos/genética , Dados de Sequência Molecular , N-Acetilglucosaminiltransferases/genética , Oligossacarídeos/genética
20.
J Biol Chem ; 287(35): 29599-609, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22745127

RESUMO

Studying genetic disorders in model organisms can provide insights into heritable human diseases. The Drosophila neurally altered carbohydrate (nac) mutant is deficient for neural expression of the HRP epitope, which consists of N-glycans with core α1,3-linked fucose residues. Here, we show that a conserved serine residue in the Golgi GDP-fucose transporter (GFR) is substituted by leucine in nac(1) flies, which abolishes GDP-fucose transport in vivo and in vitro. This loss of function is due to a biochemical defect, not to destabilization or mistargeting of the mutant GFR protein. Mass spectrometry and HPLC analysis showed that nac(1) mutants lack not only core α1,3-linked, but also core α1,6-linked fucose residues on their N-glycans. Thus, the nac(1) Gfr mutation produces a previously unrecognized general defect in N-glycan core fucosylation. Transgenic expression of a wild-type Gfr gene restored the HRP epitope in neural tissues, directly demonstrating that the Gfr mutation is solely responsible for the neural HRP epitope deficiency in the nac(1) mutant. These results validate the Drosophila nac(1) mutant as a model for the human congenital disorder of glycosylation, CDG-IIc (also known as LAD-II), which is also the result of a GFR deficiency.


Assuntos
Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica , Complexo de Golgi/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Mutação , Proteínas do Tecido Nervoso/metabolismo , Tecido Nervoso/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Epitopos/genética , Epitopos/metabolismo , Glicosilação , Complexo de Golgi/genética , Humanos , Proteínas de Transporte de Monossacarídeos/genética , Proteínas do Tecido Nervoso/genética , Transporte Proteico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA