Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 517(7533): 165-169, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25567280

RESUMO

Yeasts, which have been a component of the human diet for at least 7,000 years, possess an elaborate cell wall α-mannan. The influence of yeast mannan on the ecology of the human microbiota is unknown. Here we show that yeast α-mannan is a viable food source for the Gram-negative bacterium Bacteroides thetaiotaomicron, a dominant member of the microbiota. Detailed biochemical analysis and targeted gene disruption studies support a model whereby limited cleavage of α-mannan on the surface generates large oligosaccharides that are subsequently depolymerized to mannose by the action of periplasmic enzymes. Co-culturing studies showed that metabolism of yeast mannan by B. thetaiotaomicron presents a 'selfish' model for the catabolism of this difficult to breakdown polysaccharide. Genomic comparison with B. thetaiotaomicron in conjunction with cell culture studies show that a cohort of highly successful members of the microbiota has evolved to consume sterically-restricted yeast glycans, an adaptation that may reflect the incorporation of eukaryotic microorganisms into the human diet.


Assuntos
Bacteroidetes/metabolismo , Trato Gastrointestinal/microbiologia , Mananas/metabolismo , Modelos Biológicos , Leveduras/química , Animais , Bacteroidetes/citologia , Bacteroidetes/enzimologia , Bacteroidetes/genética , Evolução Biológica , Configuração de Carboidratos , Dieta , Enzimas/genética , Enzimas/metabolismo , Feminino , Loci Gênicos/genética , Vida Livre de Germes , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Masculino , Mananas/química , Manose/metabolismo , Camundongos , Modelos Moleculares , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Periplasma/enzimologia
3.
Microb Cell Fact ; 11: 53, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22548968

RESUMO

BACKGROUND: Protein-based therapeutics represent the fastest growing class of compounds in the pharmaceutical industry. This has created an increasing demand for powerful expression systems. Yeast systems are widely used, convenient and cost-effective. Yarrowia lipolytica is a suitable host that is generally regarded as safe (GRAS). Yeasts, however, modify their glycoproteins with heterogeneous glycans containing mainly mannoses, which complicates downstream processing and often interferes with protein function in man. Our aim was to glyco-engineer Y. lipolytica to abolish the heterogeneous, yeast-specific glycosylation and to obtain homogeneous human high-mannose type glycosylation. RESULTS: We engineered Y. lipolytica to produce homogeneous human-type terminal-mannose glycosylated proteins, i.e. glycosylated with Man8GlcNAc2 or Man5GlcNAc2. First, we inactivated the yeast-specific Golgi α-1,6-mannosyltransferases YlOch1p and YlMnn9p; the former inactivation yielded a strain producing homogeneous Man8GlcNAc2 glycoproteins. We tested this strain by expressing glucocerebrosidase and found that the hypermannosylation-related heterogeneity was eliminated. Furthermore, detailed analysis of N-glycans showed that YlOch1p and YlMnn9p, despite some initial uncertainty about their function, are most likely the α-1,6-mannosyltransferases responsible for the addition of the first and second mannose residue, respectively, to the glycan backbone. Second, introduction of an ER-retained α-1,2-mannosidase yielded a strain producing proteins homogeneously glycosylated with Man5GlcNAc2. The use of the endogenous LIP2pre signal sequence and codon optimization greatly improved the efficiency of this enzyme. CONCLUSIONS: We generated a Y. lipolytica expression platform for the production of heterologous glycoproteins that are homogenously glycosylated with either Man8GlcNAc2 or Man5GlcNAc2 N-glycans. This platform expands the utility of Y. lipolytica as a heterologous expression host and makes it possible to produce glycoproteins with homogeneously glycosylated N-glycans of the human high-mannose-type, which greatly broadens the application scope of these glycoproteins.


Assuntos
Glicoproteínas/metabolismo , Yarrowia/metabolismo , Sequência de Carboidratos , Proteínas Fúngicas/genética , Engenharia Genética , Glucosilceramidase/genética , Glucosilceramidase/metabolismo , Glicoproteínas/genética , Glicosilação , Humanos , Manose/metabolismo , Manosidases/genética , Manosidases/metabolismo , Manosiltransferases/genética , Manosiltransferases/metabolismo , Dados de Sequência Molecular , Polissacarídeos/química , Polissacarídeos/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Trichoderma/enzimologia
4.
Eukaryot Cell ; 7(8): 1268-77, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18552284

RESUMO

The filamentous fungus Aspergillus fumigatus is responsible for a lethal disease called invasive aspergillosis that affects immunocompromised patients. This disease, like other human fungal diseases, is generally treated by compounds targeting the primary fungal cell membrane sterol. Recently, glucan synthesis inhibitors were added to the limited antifungal arsenal and encouraged the search for novel targets in cell wall biosynthesis. Although galactomannan is a major component of the A. fumigatus cell wall and extracellular matrix, the biosynthesis and role of galactomannan are currently unknown. By a targeted gene deletion approach, we demonstrate that UDP-galactopyranose mutase, a key enzyme of galactofuranose metabolism, controls the biosynthesis of galactomannan and galactofuranose containing glycoconjugates. The glfA deletion mutant generated in this study is devoid of galactofuranose and displays attenuated virulence in a low-dose mouse model of invasive aspergillosis that likely reflects the impaired growth of the mutant at mammalian body temperature. Furthermore, the absence of galactofuranose results in a thinner cell wall that correlates with an increased susceptibility to several antifungal agents. The UDP-galactopyranose mutase thus appears to be an appealing adjunct therapeutic target in combination with other drugs against A. fumigatus. Its absence from mammalian cells indeed offers a considerable advantage to achieve therapeutic selectivity.


Assuntos
Aspergilose/enzimologia , Aspergillus fumigatus/enzimologia , Parede Celular/metabolismo , Furanos/metabolismo , Galactose/metabolismo , Transferases Intramoleculares/metabolismo , Mananas/metabolismo , Animais , Aspergilose/genética , Aspergilose/fisiopatologia , Aspergillus fumigatus/genética , Aspergillus fumigatus/patogenicidade , Temperatura Corporal/fisiologia , Proliferação de Células , Parede Celular/genética , Modelos Animais de Doenças , Farmacorresistência Fúngica/genética , Feminino , Regulação Fúngica da Expressão Gênica/genética , Hospedeiro Imunocomprometido/fisiologia , Transferases Intramoleculares/genética , Camundongos , Camundongos Endogâmicos BALB C , Infecções Oportunistas/genética , Infecções Oportunistas/metabolismo , Infecções Oportunistas/fisiopatologia , Virulência/genética
5.
Methods Mol Biol ; 389: 119-38, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17951639

RESUMO

Glycosylation is an important issue in heterologous protein production for therapeutic applications. Glycoproteins produced in Pichia pastoris contain high mannose glycan structures that can hamper downstream processing, might be immunogenic, and cause rapid clearance from the circulation. This chapter describes a method that helps solving these glycosylation-related problems by inactivation of OCH1, overexpression of an HDEL-tagged mannosidase, and overexpression of a Kre2/GlcNAc-transferase I chimeric enzyme. Different plasmids are described as well as glycan analysis methods.


Assuntos
Plasmídeos/genética , Polissacarídeos/biossíntese , Retículo Endoplasmático/enzimologia , Glicosilação , Complexo de Golgi/enzimologia , Humanos , Manosidases/metabolismo , Manosiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Pichia/enzimologia
6.
PLoS One ; 7(6): e39976, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22768188

RESUMO

Yarrowia lipolytica is a dimorphic yeast that efficiently secretes various heterologous proteins and is classified as "generally recognized as safe." Therefore, it is an attractive protein production host. However, yeasts modify glycoproteins with non-human high mannose-type N-glycans. These structures reduce the protein half-life in vivo and can be immunogenic in man. Here, we describe how we genetically engineered N-glycan biosynthesis in Yarrowia lipolytica so that it produces Man(3)GlcNAc(2) structures on its glycoproteins. We obtained unprecedented levels of homogeneity of this glycanstructure. This is the ideal starting point for building human-like sugars. Disruption of the ALG3 gene resulted in modification of proteins mainly with Man(5)GlcNAc(2) and GlcMan(5)GlcNAc(2) glycans, and to a lesser extent with Glc(2)Man(5)GlcNAc(2) glycans. To avoid underoccupancy of glycosylation sites, we concomitantly overexpressed ALG6. We also explored several approaches to remove the terminal glucose residues, which hamper further humanization of N-glycosylation; overexpression of the heterodimeric Apergillus niger glucosidase II proved to be the most effective approach. Finally, we overexpressed an α-1,2-mannosidase to obtain Man(3)GlcNAc(2) structures, which are substrates for the synthesis of complex-type glycans. The final Yarrowia lipolytica strain produces proteins glycosylated with the trimannosyl core N-glycan (Man(3)GlcNAc(2)), which is the common core of all complex-type N-glycans. All these glycans can be constructed on the obtained trimannosyl N-glycan using either in vivo or in vitro modification with the appropriate glycosyltransferases. The results demonstrate the high potential of Yarrowia lipolytica to be developed as an efficient expression system for the production of glycoproteins with humanized glycans.


Assuntos
Engenharia Genética , Glicoproteínas/biossíntese , Oligossacarídeos/biossíntese , Polissacarídeos/biossíntese , Yarrowia/genética , Animais , Eletroforese em Gel de Poliacrilamida , Retículo Endoplasmático/enzimologia , Técnicas de Inativação de Genes , Genes Fúngicos/genética , Glucose/metabolismo , Glicoproteínas/química , Glicosilação , Humanos , Lipase/metabolismo , Manosidases/metabolismo , Oligossacarídeos/química , Polissacarídeos/química , Ratos , Trypanosoma brucei brucei/enzimologia , Yarrowia/enzimologia , alfa-Glucosidases/metabolismo
7.
Nat Biotechnol ; 30(12): 1225-31, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23159880

RESUMO

Lysosomal storage diseases are treated with human lysosomal enzymes produced in mammalian cells. Such enzyme therapeutics contain relatively low levels of mannose-6-phosphate, which is required to target them to the lysosomes of patient cells. Here we describe a method for increasing mannose-6-phosphate modification of lysosomal enzymes produced in yeast. We identified a glycosidase from C. cellulans that 'uncaps' N-glycans modified by yeast-type mannose-Pi-6-mannose to generate mammalian-type N-glycans with a mannose-6-phosphate substitution. Determination of the crystal structure of this glycosidase provided insight into its substrate specificity. We used this uncapping enzyme together with α-mannosidase to produce in yeast a form of the Pompe disease enzyme α-glucosidase rich in mannose-6-phosphate. Compared with the currently used therapeutic version, this form of α-glucosidase was more efficiently taken up by fibroblasts from Pompe disease patients, and it more effectively reduced cardiac muscular glycogen storage in a mouse model of the disease.


Assuntos
Glicosídeo Hidrolases/metabolismo , Lisossomos/enzimologia , Manosefosfatos/metabolismo , Animais , Arthrobacter/enzimologia , Arthrobacter/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Transporte Biológico Ativo , Biotecnologia , Domínio Catalítico/genética , Modelos Animais de Doenças , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Doença de Depósito de Glicogênio Tipo II/enzimologia , Doença de Depósito de Glicogênio Tipo II/genética , Glicosídeo Hidrolases/química , Glicosídeo Hidrolases/genética , Humanos , Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Doenças por Armazenamento dos Lisossomos/enzimologia , Doenças por Armazenamento dos Lisossomos/genética , Camundongos , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Pichia/enzimologia , Pichia/genética , Conformação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Yarrowia/enzimologia , Yarrowia/genética , alfa-Glucosidases/deficiência , alfa-Glucosidases/genética , alfa-Glucosidases/metabolismo
8.
Nat Protoc ; 4(1): 58-70, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19131957

RESUMO

Here we provide a protocol for engineering the N-glycosylation pathway of the yeast Pichia pastoris. The general strategy consists of the disruption of an endogenous glycosyltransferase gene (OCH1) and the stepwise introduction of heterologous glycosylation enzymes. Each engineering step results in the introduction of one glycosidase or glycosyltransferase activity into the Pichia endoplasmic reticulum or Golgi complex and consists of a number of stages: transformation with the appropriate GlycoSwitch vector, small-scale cultivation of a number of transformants, sugar analysis and heterologous protein expression analysis. If desired, the resulting clone can be further engineered by repeating the procedure with the next GlycoSwitch vector. Each engineering step takes approximately 3 weeks. The conversion of any wild-type Pichia strain into a strain that modifies its glycoproteins with Gal(2)GlcNAc(2)Man(3)GlcNAc(2)N-glycans requires the introduction of five GlycoSwitch vectors. Three examples of the full engineering procedure are provided to illustrate the results that can be expected.


Assuntos
Glucosiltransferases/genética , Pichia/genética , Engenharia de Proteínas/métodos , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/metabolismo , Vetores Genéticos/genética , Glucosiltransferases/metabolismo , Glicosilação , Complexo de Golgi/enzimologia , Complexo de Golgi/metabolismo , Pichia/metabolismo
9.
Protein Expr Purif ; 53(2): 275-82, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17317217

RESUMO

The Nicotiana tabacum lectin, also called Nictaba, is a nucleocytoplasmic plant lectin expressed in tobacco leaves after exposure to jasmonates. Purification of the lectin from raw material is a time-consuming process, demanding large amounts of induced plant material. In addition, the lectin yield is low and purified lectin fractions are always contaminated with low molecular weight compounds such as phenols. In a way to improve and facilitate the purification of the tobacco lectin, we cloned the Nictaba gene in a vector optimized for protein expression in the methylotrophic yeast Pichia pastoris. In this report, we present data of the expression profile of recombinant Nictaba in the P. pastoris culture medium and in P. pastoris cells together with the purification strategy using ion exchange chromatography and affinity chromatography on a column with immobilized ovomucoid. Pichia transformants were estimated to express approximately 6mg of recombinant lectin per liter medium after a 72h culture. SDS-PAGE and Western blot analysis revealed that the recombinant lectin expressed in Pichia exists in two molecular forms. Edman degradation and mass spectrometry analysis confirmed the presence of at least two forms of recombinant lectin with molecular weights of 19,060 and 20,100Da, corresponding to lectin polypeptides similar to the fully processed Nictaba which is N-terminally blocked, and Nictaba extended at the N-terminus with the amino acids residues EAEAYVEFT due to incomplete processing of the alpha-factor mating sequence. Further characterisation of the recombinant lectin revealed agglutination and carbohydrate-binding properties similar to the native tobacco lectin.


Assuntos
Nicotiana/genética , Pichia/genética , Lectinas de Plantas/genética , Lectinas de Plantas/isolamento & purificação , Sequência de Aminoácidos , Sequência de Bases , Cromatografia de Afinidade , Cromatografia por Troca Iônica , Clonagem Molecular , DNA de Plantas/genética , Perfilação da Expressão Gênica , Genes de Plantas , Vetores Genéticos , Dados de Sequência Molecular , Plasmídeos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Transformação Genética
10.
J Gastroenterol Hepatol ; 22(7): 1148-54, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17608861

RESUMO

BACKGROUND: Non-invasive staging of human liver fibrosis is a desirable objective that remains under extensive evaluation. Animal model systems are often used for studying human liver disease and screening antifibrotic compounds. The aim of the present study was to investigate the potential use of serum N-glycan profiles to evaluate liver fibrosis in a rat model. METHODS: Liver fibrosis and cirrhosis were induced in rats by oral administration of CCl(4). Liver injury was assessed biochemically (alanine aminotransferase [ALT] activity, aspartate aminotransferase [AST] activity and total bilirubin) and histologically. The N-glycan profile (GlycoTest) was performed using DNA sequencer-assisted-fluorophore-assisted carbohydrate electrophoresis technology. In parallel, the effect of cotreatment with antifibrotic interferon-gamma (IFN-gamma) was studied. RESULTS: The biopsy scoring system showed that CCl(4) induced early fibrosis (F < 1-2) in rats after 3 weeks of treatment, and cirrhosis (F4) after 12 weeks. Significant increases in ALT activity, AST activity and total bilirubin levels were detected only after 12 weeks of CCl(4) treatment. GlycoTest showed three glycans were significantly altered in the CCl(4)-goup. Peak 3 started at week 6, at an early stage in fibrosis development (F < 1-2), whereas peaks 4 and 5 occurred at week 9, at which time mild liver fibrosis (F = 1-2) had developed. The changes in the CCl(4)-IFN-gamma group were intermediate between the CCl(4)- and the control groups. CONCLUSION: The GlycoTest is much more sensitive than biochemical tests for evaluating liver fibrosis/cirrhosis in the rat model. The test can also be used as a non-invasive marker for screening and monitoring the antifibrotic activity of potential therapeutic compounds.


Assuntos
Cirrose Hepática/sangue , Polissacarídeos/sangue , Animais , Tetracloreto de Carbono/administração & dosagem , Cirrose Hepática/induzido quimicamente , Masculino , Ratos , Ratos Wistar
11.
Glycobiology ; 17(1): 36-45, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17000699

RESUMO

MFECP1 is a mannosylated antibody-enzyme fusion protein used in antibody-directed enzyme prodrug therapy (ADEPT). The antibody selectively targets tumor cells and the targeted enzyme converts a prodrug into a toxic drug. MFECP1 is obtained from expression in the yeast Pichia pastoris and produced to clinical grade. The P. pastoris-derived mannosylation of the fusion protein aids rapid normal tissue clearance required for successful ADEPT. The work presented provides evidence that MFECP1 is cleared by the endocytic and phagocytic mannose receptor (MR), which is known to bind to mannose-terminating glycans. MR-transfected fibroblast cells internalize MFECP1 as revealed by flow cytometry and confocal microscopy. Immunofluorescence microscopy shows that in vivo clearance in mice occurs predominantly by MR on liver sinusoidal endothelial cells, although MR is also expressed on adjacent Kupffer cells. In the spleen, MFECP1 is taken up by MR-expressing macrophages residing in the red pulp and not by dendritic cells which are found in the marginal zone and white pulp. Clearance can be inhibited in vivo by the MR inhibitor mannan as shown by increased enzyme activities in blood. The work improves understanding of interactions of MFECP1 with normal tissue, shows that glycosylation can be exploited in the design of recombinant anticancer therapeutics and opens the ways for optimizing pharmacokinetics of mannosylated recombinant therapeutics.


Assuntos
Imunoconjugados/farmacocinética , Imunoconjugados/uso terapêutico , Neoplasias Experimentais/terapia , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Antígeno Carcinoembrionário/imunologia , Células Cultivadas , Imunoconjugados/química , Imunoterapia/métodos , Lectinas Tipo C/metabolismo , Fígado/química , Mananas/farmacologia , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Taxa de Depuração Metabólica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Polissacarídeos/análise , Pró-Fármacos/síntese química , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Pró-Fármacos/uso terapêutico , Ratos , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes de Fusão/química , Baço/química , Distribuição Tecidual , Transfecção
12.
Appl Environ Microbiol ; 70(5): 2639-46, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15128513

RESUMO

The Pichia pastoris N-glycosylation pathway is only partially homologous to the pathway in human cells. In the Golgi apparatus, human cells synthesize complex oligosaccharides, whereas Pichia cells form mannose structures that can contain up to 40 mannose residues. This hypermannosylation of secreted glycoproteins hampers the downstream processing of heterologously expressed glycoproteins and leads to the production of protein-based therapeutic agents that are rapidly cleared from the blood because of the presence of terminal mannose residues. Here, we describe engineering of the P. pastoris N-glycosylation pathway to produce nonhyperglycosylated hybrid glycans. This was accomplished by inactivation of OCH1 and overexpression of an alpha-1,2-mannosidase retained in the endoplasmic reticulum and N-acetylglucosaminyltransferase I and beta-1,4-galactosyltransferase retained in the Golgi apparatus. The engineered strain synthesized a nonsialylated hybrid-type N-linked oligosaccharide structure on its glycoproteins. The procedures which we developed allow glycan engineering of any P. pastoris expression strain and can yield up to 90% homogeneous protein-linked oligosaccharides.


Assuntos
Engenharia Genética/métodos , Pichia/metabolismo , Polissacarídeos/biossíntese , Biotecnologia/métodos , Retículo Endoplasmático/enzimologia , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Glicosilação , Complexo de Golgi/enzimologia , Humanos , Manosidases/genética , Manosidases/metabolismo , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Pichia/genética , Pichia/crescimento & desenvolvimento , Polissacarídeos/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA