Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Microb Cell Fact ; 21(1): 124, 2022 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-35729556

RESUMO

Replacement of petrochemical-based materials with microbially produced biodegradable alternatives calls for industrially attractive fermentation processes. Lignocellulosic materials offer non-edible alternatives for cultivated sugars, but require often use of expensive sugar releasing enzymes, such as ß-glucosidases. These cellulose treatment costs could be reduced if microbial production hosts could use short cellodextrins such as cellobiose directly as their substrates. In this study, we demonstrate production of poly(hydroxybutyrate) (PHB) in yeast Saccharomyces cerevisiae using cellobiose as a sole carbon source. Yeast strains expressing PHB pathway genes from Cupriavidus necator and cellodextrin transporter gene CDT-1 from Neurospora crassa were complemented either with ß-glucosidase gene GH1-1 from N. crassa or with cellobiose phosphorylase gene cbp from Ruminococcus flavefaciens. These cellobiose utilization routes either with Gh1-1 or Cbp enzymes differ in energetics and dynamics. However, both routes enabled higher PHB production per consumed sugar and higher PHB accumulation % of cell dry weight (CDW) than use of glucose as a carbon source. As expected, the strains with Gh1-1 consumed cellobiose faster than the strains with Cbp, both in flask and bioreactor batch cultures. In shake flasks, higher final PHB accumulation % of CDW was reached with Cbp route (10.0 ± 0.3%) than with Gh1-1 route (8.1 ± 0.2%). However, a higher PHB accumulation was achieved in better aerated and pH-controlled bioreactors, in comparison to shake flasks, and the relative performance of strains switched. In bioreactors, notable PHB accumulation levels per CDW of 13.4 ± 0.9% and 18.5 ± 3.9% were achieved with Cbp and Gh1-1 routes, respectively. The average molecular weights of accumulated PHB were similar using both routes; approximately 500 kDa and 450 kDa for strains expressing either cbp or GH1-1 genes, respectively. The formation of PHB with high molecular weights, combined with efficient cellobiose conversion, demonstrates a highly potential solution for improving attractiveness of sustainable polymer production using microbial cells.


Assuntos
Celobiose , Saccharomyces cerevisiae , Carbono/metabolismo , Celobiose/metabolismo , Fermentação , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , beta-Glucosidase/metabolismo
2.
FEMS Yeast Res ; 20(3)2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32310262

RESUMO

Processed lignocellulosic biomass is a source of mixed sugars that can be used for microbial fermentation into fuels or higher value products, like chemicals. Previously, the yeast Saccharomyces cerevisiae was engineered to utilize its cellodextrins through the heterologous expression of sugar transporters together with an intracellular expressed ß-glucosidase. In this study, we screened a selection of eight (putative) cellodextrin transporters from different yeast and fungal hosts in order to extend the catalogue of available cellobiose transporters for cellobiose fermentation in S. cerevisiae. We confirmed that several in silico predicted cellodextrin transporters from Aspergillus niger were capable of transporting cellobiose with low affinity. In addition, we found a novel cellobiose transporter from the yeast Lipomyces starkeyi, encoded by the gene Ls120451. This transporter allowed efficient growth on cellobiose, while it also grew on glucose and lactose, but not cellotriose nor cellotetraose. We characterized the transporter more in-depth together with the transporter CdtG from Penicillium oxalicum. CdtG showed to be slightly more efficient in cellobiose consumption than Ls120451 at concentrations below 1.0 g/L. Ls120451 was more efficient in cellobiose consumption at higher concentrations and strains expressing this transporter grew slightly slower, but produced up to 30% more ethanol than CdtG.


Assuntos
Celobiose/metabolismo , Fermentação , Lipomyces/genética , Proteínas de Membrana Transportadoras/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transporte Biológico , Biomassa , Celulose/análogos & derivados , Celulose/metabolismo , Dextrinas/metabolismo , Etanol/metabolismo , Lipomyces/crescimento & desenvolvimento , Lipomyces/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Penicillium/genética
3.
Biomacromolecules ; 21(10): 4355-4364, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-32960595

RESUMO

In nature, various organisms produce cellulose as microfibrils, which are processed into their nano- and microfibrillar and/or crystalline components by humans in order to obtain desired material properties. Interestingly, the natural synthesis machinery can be circumvented by enzymatically synthesizing cellulose from precursor molecules in vitro. This approach is appealing for producing tailor-made cellulosic particles and materials because it enables optimization of the reaction conditions for cellulose synthesis in order to generate particles with a desired morphology in their pure form. Here, we present enzymatic cellulose synthesis catalyzed by the reverse reaction of Clostridium thermocellum cellodextrin phosphorylase in vitro. We were able to produce cellulose II nanofibril networks in all conditions tested, using varying concentrations of the glycosyl acceptors d-glucose or d-cellobiose (0.5, 5, and 50 mM). We show that shorter cellulose chains assemble into flat ribbon-like fibrils with greater diameter, while longer chains assemble into cylindrical fibrils with smaller diameter.


Assuntos
Celulose , Clostridium thermocellum , Glucosiltransferases , Catálise , Nanofibras
4.
Yeast ; 35(4): 331-341, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29159981

RESUMO

The cellular changes induced by heterologous protein expression in the yeast Saccharomyces cerevisiae have been analysed on many levels and found to be significant. However, even though high-level protein production poses a metabolic burden, evaluation of the expression host at the level of the metabolome has often been neglected. We present a comparison of metabolite profiles of a wild-type strain with those of three strains producing recombinant antibody variants of increasing size and complexity: an scFv fragment, an scFv-Fc fusion protein and a full-length IgG molecule. Under producing conditions, all three recombinant strains showed a clear decrease in growth rate compared with the wild-type strain and the severity of the growth phenotype increased with size of the protein. The levels of 76 intracellular metabolites were determined using a targeted (semi) quantitative mass spectrometry based approach. Based on unsupervised and supervised multivariate analysis of metabolite profiles, together with pathway activity profiling, the recombinant strains were found to be significantly different from each other and from the wild-type strain. We observed the most prominent changes in metabolite levels for metabolites involved in amino acid and redox metabolism. Induction of the unfolded protein response was detected in all producing strains and is considered to be a contributing factor to the overall metabolic burden on the cells.


Assuntos
Anticorpos/metabolismo , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/metabolismo , Aminoácidos/metabolismo , Reatores Biológicos , Metabolismo Energético/fisiologia , Redes e Vias Metabólicas , Metaboloma , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/imunologia
5.
FEMS Yeast Res ; 17(1)2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27956492

RESUMO

Cost-effective manufacturing of biopharmaceuticals in non-mammalian hosts still requires tremendous efforts in strain development. In order to expedite identification of novel leads for strain engineering, we used a transposon-mutagenized yeast genomic DNA library to create a collection of Saccharomyces cerevisiae deletion strains expressing a full-length IgG antibody. Using a high-throughput screening, transformants with either significantly higher or lower IgG expression were selected. The integration site of the transposon in three of the selected strains was located by DNA sequencing. The inserted DNA lay within the VPS30 and TAR1 open reading frame, and upstream of the HEM13 open reading frame. The complete coding sequence of these genes was deleted in the wild-type strain background to confirm the IgG expression phenotypes. Production of recombinant antibody was increased 2-fold in the Δvps30 strain, but only mildly affected secretion levels in the Δtar1 strain. Remarkably, expression of endogenous yeast acid phosphatase was increased 1.7- and 2.4-fold in Δvps30 and Δtar1 strains. The study confirmed the power of genome-wide high-throughput screens for strain development and highlights the importance of using the target molecule during the screening process.


Assuntos
Deleção de Genes , Genes Fúngicos , Imunoglobulina G/biossíntese , Proteínas Recombinantes/biossíntese , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Elementos de DNA Transponíveis , Testes Genéticos , Imunoglobulina G/genética , Microbiologia Industrial/métodos , Engenharia Metabólica/métodos , Mutagênese Insercional , Proteínas Recombinantes/genética
6.
Glycoconj J ; 33(2): 189-99, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26983412

RESUMO

N-glycosylation is an important feature of therapeutic and other industrially relevant proteins, and engineering of the N-glycosylation pathway provides opportunities for developing alternative, non-mammalian glycoprotein expression systems. Among yeasts, Saccharomyces cerevisiae is the most established host organism used in therapeutic protein production and therefore an interesting host for glycoengineering. In this work, we present further improvements in the humanization of the N-glycans in a recently developed S. cerevisiae strain. In this strain, a tailored trimannosyl lipid-linked oligosaccharide is formed and transferred to the protein, followed by complex-type glycan formation by Golgi apparatus-targeted human N-acetylglucosamine transferases. We improved the glycan pattern of the glycoengineered strain both in terms of glycoform homogeneity and the efficiency of complex-type glycosylation. Most of the interfering structures present in the glycoengineered strain were eliminated by deletion of the MNN1 gene. The relative abundance of the complex-type target glycan was increased by the expression of a UDP-N-acetylglucosamine transporter from Kluyveromyces lactis, indicating that the import of UDP-N-acetylglucosamine into the Golgi apparatus is a limiting factor for efficient complex-type N-glycosylation in S. cerevisiae. By a combination of the MNN1 deletion and the expression of a UDP-N-acetylglucosamine transporter, a strain forming complex-type glycans with a significantly improved homogeneity was obtained. Our results represent a further step towards obtaining humanized glycoproteins with a high homogeneity in S. cerevisiae.


Assuntos
Polissacarídeos Fúngicos/biossíntese , Oligossacarídeos/biossíntese , Saccharomyces cerevisiae/metabolismo , Configuração de Carboidratos , Polissacarídeos Fúngicos/química , Polissacarídeos Fúngicos/genética , Deleção de Genes , Glicosilação , Humanos , Kluyveromyces/genética , Kluyveromyces/metabolismo , Manosiltransferases/genética , Manosiltransferases/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Oligossacarídeos/química , Oligossacarídeos/genética , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
7.
Microb Cell Fact ; 15: 87, 2016 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-27216259

RESUMO

BACKGROUND: The yeast Saccharomyces cerevisiae provides intriguing possibilities for synthetic biology and bioprocess applications, but its use is still constrained by cellular characteristics that limit the product yields. Considering the production of advanced biopharmaceuticals, a major hindrance lies in the yeast endoplasmic reticulum (ER), as it is not equipped for efficient and large scale folding of complex proteins, such as human antibodies. RESULTS: Following the example of professional secretory cells, we show that inducing an ER expansion in yeast by deleting the lipid-regulator gene OPI1 can improve the secretion capacity of full-length antibodies up to fourfold. Based on wild-type and ER-enlarged yeast strains, we conducted a screening of a folding factor overexpression library to identify proteins and their expression levels that enhance the secretion of antibodies. Out of six genes tested, addition of the peptidyl-prolyl isomerase CPR5 provided the most beneficial effect on specific product yield while PDI1, ERO1, KAR2, LHS1 and SIL1 had a mild or even negative effect to antibody secretion efficiency. Combining genes for ER enhancement did not induce any significant additional effect compared to addition of just one element. By combining the Δopi1 strain, with the enlarged ER, with CPR5 overexpression, we were able to boost the specific antibody product yield by a factor of 10 relative to the non-engineered strain. CONCLUSIONS: Engineering protein folding in vivo is a major task for biopharmaceuticals production in yeast and needs to be optimized at several levels. By rational strain design and high-throughput screening applications we were able to increase the specific secreted antibody yields of S. cerevisiae up to 10-fold, providing a promising strain for further process optimization and platform development for antibody production.


Assuntos
Anticorpos/metabolismo , Retículo Endoplasmático/metabolismo , Saccharomyces cerevisiae/metabolismo , Anticorpos/química , Anticorpos/genética , Peptidilprolil Isomerase/genética , Peptidilprolil Isomerase/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Dobramento de Proteína , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Temperatura
8.
Appl Microbiol Biotechnol ; 99(21): 9061-71, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26184977

RESUMO

One of the main limitations for heterologous protein production in the yeast Saccharomyces cerevisiae is the protein-folding capacity in the endoplasmic reticulum (ER). Accumulation of unfolded proteins triggers the unfolded protein response (UPR), which resolves the stress by increasing the capacity for protein folding and removal of unfolded proteins by the ER-associated degradation (ERAD) system. In order to analyze the influence of ERAD on production of a human IgG, we disrupted ERAD at different stages by deletion of the HTM1, YOS9, HRD1, HRD3, or UBC7 gene, with or without a disruption of the UPR by deletion of the IRE1 gene. All deletion strains were viable and did not exhibit a growth phenotype under normal growth conditions. Deletion of HTM1 resulted in a small increase in antibody production, whereas a small decrease in antibody production was observed in the Δhrd1, Δhrd3, and Δubc7 yeast strains, and a stronger decrease in the Δyos9 yeast strain. Deletion of the IRE1 gene had contrasting effects in the ERAD mutants, with a strongly decreased production in wild-type cells and partially reversed effects in combination with the Δhtm1 or the Δyos9 deletions. In order to study IgG clearance from the ER, an assay was developed using the inhibitory effect of glucose on the GAL1 promoter that is driving IgG expression. The Δyos9Δire1and Δhtm1Δire1 strains showed a delayed IgG clearance from the cells, showing that removal of components for the generation and recognition of the glycan signal needed for ERAD-mediated protein degradation might increase the IgG ER residence time.


Assuntos
Retículo Endoplasmático/metabolismo , Imunoglobulina G/metabolismo , Dobramento de Proteína , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/metabolismo , Deleção de Genes , Humanos , Imunoglobulina G/genética , Viabilidade Microbiana , Proteólise , Proteínas Recombinantes/genética , Saccharomyces cerevisiae/genética , Resposta a Proteínas não Dobradas
9.
Eur Food Res Technol ; : 1-10, 2023 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-37362347

RESUMO

The so-called "craft beer revolution" has increased the demand for new styles of beers, often with new ingredients like flavour extracts. In recent years, synthetic biology has realized the production of a plethora of plant secondary metabolites in microbial hosts, which could provide an alternative source for these compounds. In this study, we selected a in situ flavour production approach for grape flavour addition. We used an O-methyl anthranilate (OmANT) producing laboratory Saccharomyces cerevisiae strain in co-fermentations with an industrial beer yeast strain WLP644. The laboratory strain provided an ease of genetic manipulation and the desirable properties of the WLP644 strain were not modified in this approach. In shake flasks, a 10:90 ratio of the yeasts produced grape flavoured beer with the yeast produced flavour compound in a range normally used for flavoured beverages. Hopped and unhopped beers were analysed by VTT's trained sensory panel and with olfactory GC-MS. OmANT was successfully detected from the beers as a floral odour and flavour. Moreover, no off-flavours were detected and aroma profiles outside the grape flavour were rather similar. These results indicate that the co-fermentation principle is a suitable approach to change the flavour profiles of beers with a simple yeast strain drop-in approach. Supplementary Information: The online version contains supplementary material available at 10.1007/s00217-023-04274-1.

10.
Biotechnol J ; 12(8)2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28429845

RESUMO

Therapeutic protein production in yeast is a reality in industry with an untapped potential to expand to more complex proteins, such as full-length antibodies. Despite numerous engineering approaches, cellular limitations are preventing the use of Saccharomyces cerevisiae as the titers of recombinant antibodies are currently not competitive. Instead of a host specific approach, the possibility of adopting the features from native producers of antibodies, plasma cells, to improve antibody production in yeast. A subset of mammalian folding factors upregulated in plasma cells for expression in yeast and screened for beneficial effects on antibody secretion using a high-throughput ELISA platform was selected. Co-expression of the mammalian chaperone BiP, the co-chaperone GRP170, or the peptidyl-prolyl isomerase FKBP2, with the antibody improved specific product yields up to two-fold. By comparing strains expressing FKBP2 or the yeast PPIase Cpr5p, the authors demonstrate that speeding up peptidyl-prolyl isomerization by upregulation of catalyzing enzymes is a key factor to improve antibody titers in yeast. The findings show that following the route of plasma cells can improve product titers and contribute to developing an alternative yeast-based antibody factory.


Assuntos
Anticorpos/genética , Formação de Anticorpos/genética , Proteínas Recombinantes/biossíntese , Saccharomyces cerevisiae/genética , Anticorpos/imunologia , Formação de Anticorpos/imunologia , Chaperona BiP do Retículo Endoplasmático , Glicoproteínas/biossíntese , Glicoproteínas/genética , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico/biossíntese , Proteínas de Choque Térmico/genética , Peptidilprolil Isomerase/biossíntese , Peptidilprolil Isomerase/genética , Plasmócitos/imunologia , Plasmócitos/metabolismo , Dobramento de Proteína , Proteínas Recombinantes/genética , Saccharomyces cerevisiae/química
11.
N Biotechnol ; 31(6): 532-7, 2014 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-24632452

RESUMO

N-linked glycosylation of proteins is one of the most common posttranslational modifications. N-glycan structures and N-glycosylation efficiency are crucial parameters in the production of N-glycosylated proteins. Yeast cells can be seen as an attractive production host for therapeutic glycoproteins and pioneering work of glycoengineering was performed in Pichia pastoris, realizing yeast strains capable of producing defined, human-type N-glycans. Most strategies used for glycoengineering rely on the modification of the lipid-linked oligosaccharide biosynthesis for the generation of the substrate for Golgi-localized glycosyltransferases. However, modifications in the lipid-linked oligosaccharide biosynthesis often result in the accumulation of intermediate structures and cause hypoglycosylation of client proteins. In order to ensure complete N-glycosylation, the flow of lipid-linked oligosaccharide through the biosynthetic pathway and the transfer of the oligosaccharide from the donor lipid onto the protein have to be optimized. A promising tool to improve site occupancy is the expression of protozoan oligosaccharyltransferases, which possess altered specificities for the oligosaccharide and also for the protein acceptor site. Furthermore, flipping of the lipid-linked oligosaccharide into the ER lumen can be improved by overexpression of an artificial flippase. Improving the glycosylation efficiency ensures that not only homogeneous N-glycan structures are generated, but also client proteins are fully glycosylated.


Assuntos
Metabolismo dos Carboidratos , Pichia/metabolismo , Glicosilação
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa