Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Biotechnol Bioeng ; 117(4): 1172-1186, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31814104

RESUMO

Large scale biopharmaceutical production of biologics relies on the overexpression of foreign proteins by cells cultivated in stirred tank bioreactors. It is well recognized and documented fact that protein overexpression may impact host cell metabolism and that factors associated with large scale culture, such as the hydrodynamic forces and inhomogeneities within the bioreactors, may promote cellular stress. The metabolic adaptations required to support the high-level expression of recombinant proteins include increased energy production and improved secretory capacity, which, in turn, can lead to a rise of reactive oxygen species (ROS) generated through the respiration metabolism and the interaction with media components. Oxidative stress is defined as the imbalance between the production of free radicals and the antioxidant response within the cells. Accumulation of intracellular ROS can interfere with the cellular activities and exert cytotoxic effects via the alternation of cellular components. In this context, strategies aiming to alleviate oxidative stress generated during the culture have been developed to improve cell growth, productivity, and reduce product microheterogeneity. In this review, we present a summary of the different approaches used to decrease the oxidative stress in Chinese hamster ovary cells and highlight media development and cell engineering as the main pathways through which ROS levels may be kept under control.


Assuntos
Antioxidantes , Células CHO , Engenharia Celular/métodos , Estresse Oxidativo , Proteínas Recombinantes/metabolismo , Animais , Reatores Biológicos , Técnicas de Cultura de Células , Cricetinae , Cricetulus , Meios de Cultura
2.
Metab Eng Commun ; 9: e00097, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31720213

RESUMO

Biopharmaceutical industrial processes are based on high yielding stable recombinant Chinese Hamster Ovary (CHO) cells that express monoclonal antibodies. However, the process and feeding regimes need to be adapted for each new cell line, as they all have a slightly different metabolism and product performance. A main limitation for accelerating process development is that the metabolic pathways underlying this physiological variability are not yet fully understood. This study describes the evolution of intracellular fluxes during the process for 4 industrial cell lines, 2 high producers and 2 low producers (n = 3), all of them producing a different antibody. In order to understand from a metabolic point of view the phenotypic differences observed, and to find potential targets for improving specific productivity of low producers, the analysis was supported by a tailored genome-scale model and was validated with enzymatic assays performed at different days of the process. A total of 59 reactions were examined from different key pathways, namely glycolysis, pentose phosphate pathway, TCA cycle, lipid metabolism, and oxidative phosphorylation. The intracellular fluxes did not show a metabolic correlation between high producers, but the degree of similitude observed between cell lines could be confirmed with additional experimental observations. The whole analysis led to a better understanding of the metabolic requirements for all the cell lines, allowed to the identification of metabolic bottlenecks and suggested targets for further cell line engineering. This study is a successful application of a curated genome-scale model to multiple industrial cell lines, which makes the metabolic model suitable for process platform.

3.
Metab Eng ; 56: 120-129, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31526854

RESUMO

Chinese hamster ovary (CHO) cells are the preferred host for producing biopharmaceuticals. Amino acids are biologically important precursors for CHO metabolism; they serve as building blocks for proteogenesis, including synthesis of biomass and recombinant proteins, and are utilized for growth and cellular maintenance. In this work, we studied the physiological impact of disrupting a range of amino acid catabolic pathways in CHO cells. We aimed to reduce secretion of growth inhibiting metabolic by-products derived from amino acid catabolism including lactate and ammonium. To achieve this, we engineered nine genes in seven different amino acid catabolic pathways using the CRISPR-Cas9 genome editing system. For identification of target genes, we used a metabolic network reconstruction of amino acid catabolism to follow transcriptional changes in response to antibody production, which revealed candidate genes for disruption. We found that disruption of single amino acid catabolic genes reduced specific lactate and ammonium secretion while specific growth rate and integral of viable cell density were increased in many cases. Of particular interest were Hpd and Gad2 disruptions, which show unchanged AA uptake rates, while having growth rates increased up to 19%, and integral of viable cell density as much as 50% higher, and up to 26% decrease in specific ammonium production and to a lesser extent (up to 22%) decrease in lactate production. This study demonstrates the broad potential of engineering amino acid catabolism in CHO cells to achieve improved phenotypes for bioprocessing.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Reprogramação Celular , Edição de Genes , Redes e Vias Metabólicas/genética , Animais , Células CHO , Cricetulus
4.
J Biotechnol ; 306: 24-31, 2019 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-31465797

RESUMO

In recombinant protein expression using Chinese hamster ovary (CHO) cells, chemically defined media contain essential amino acids such as branched chain amino acids (BCAAs) leucine, isoleucine and valine. Availability of amino acids is critical as these are building blocks for protein synthesis. However, breakdown of amino acids can lead to build up of toxic intermediates and metabolites that decrease cell growth, productivity and product quality. BCAA catabolism also hampers the usage of BCAAs for protein synthesis. In this work we studied the effects of disrupting the genes responsible for the first step of BCAA catabolism: branched chain aminotransferase 1 (Bcat1) and branched chain aminotransferase 2 (Bcat2). We evaluated the effect of disrupting the genes individually and in combination, and examined the effects in producer and non-producer host cells. Our experiments show that Bcat1 disruption improves cell growth in producer cells, but not in non-producers. Conversely, Bcat2 has a minor negative effect on growth in producer cells, and none in non-producers. Combined Bcat1 and Bcat2 disruption improves growth in producer cells. By-product metabolism is cell line-, clone- and producer-dependent. Overall, our results show that the effects of targeting Bcat1 and/or Bcat2 are cell line-dependent, and seemingly linked to the burden of recombinant protein expression.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Transaminases/metabolismo , Animais , Células CHO , Proliferação de Células , Sobrevivência Celular , Cricetulus , Meios de Cultura/metabolismo , Mutação , Biossíntese de Proteínas , Transaminases/genética
5.
Biotechnol Bioeng ; 116(10): 2778-2796, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31237682

RESUMO

The number of approved biopharmaceuticals, where product quality attributes remain of major importance, is increasing steadily. Within the available variety of expression hosts, the production of biopharmaceuticals faces diverse limitations with respect to posttranslational modifications (PTM), while different biopharmaceuticals demand different forms and specifications of PTMs for proper functionality. With the growing toolbox of genetic engineering technologies, it is now possible to address general as well as host- or biopharmaceutical-specific product quality obstacles. In this review, we present diverse expression systems derived from mammalians, bacteria, yeast, plants, and insects as well as available genetic engineering tools. We focus on genes for knockout/knockdown and overexpression for meaningful approaches to improve biopharmaceutical PTMs and discuss their applicability as well as future trends in the field.


Assuntos
Produtos Biológicos , Engenharia Genética , Animais , Produtos Biológicos/química , Produtos Biológicos/metabolismo , Humanos
6.
ACS Synth Biol ; 8(4): 758-774, 2019 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-30807689

RESUMO

Many branches of biology depend on stable and predictable recombinant gene expression, which has been achieved in recent years through targeted integration of the recombinant gene into defined integration sites. However, transcriptional levels of recombinant genes in characterized integration sites are controlled by multiple components of the integrated expression cassette. Lack of readily available tools has inhibited meaningful experimental investigation of the interplay between the integration site and the expression cassette components. Here we show in a systematic manner how multiple components contribute to final net expression of recombinant genes in a characterized integration site. We develop a CRISPR/Cas9-based toolbox for construction of mammalian cell lines with targeted integration of a landing pad, containing a recombinant gene under defined 5' proximal regulatory elements. Generated site-specific recombinant cell lines can be used in a streamlined recombinase-mediated cassette exchange for fast screening of different expression cassettes. Using the developed toolbox, we show that different 5' proximal regulatory elements generate distinct and robust recombinant gene expression patterns in defined integration sites of CHO cells with a wide range of transcriptional outputs. This approach facilitates the generation of user-defined and product-specific gene expression patterns for programmable mammalian cell engineering.


Assuntos
Expressão Gênica/genética , Mamíferos/genética , Proteínas Recombinantes/genética , Animais , Células CHO , Sistemas CRISPR-Cas/genética , Engenharia Celular/métodos , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Cricetulus , Recombinases/genética , Sequências Reguladoras de Ácido Nucleico/genética , Transcrição Gênica/genética
7.
Metab Eng ; 51: 9-19, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30227251

RESUMO

CHO cells have become the favorite expression system for large scale production of complex biopharmaceuticals. However, industrial strategies for upstream process development are based on empirical results, due to a lack of fundamental understanding of intracellular activities. Genome scale models of CHO cells have been reconstructed to provide an economical way of analyzing and interpreting large-omics datasets, since they add cellular context to the data. Here the most recently available CHO-DG44 genome-scale specific model was manually curated and tailored to the metabolic profile of cell lines used for industrial protein production, by modifying 601 reactions. Generic changes were applied to simplify the model and cope with missing constraints related to regulatory effects as well as thermodynamic and osmotic forces. Cell line specific changes were related to the metabolism of high-yielding production cell lines. The model was semi-constrained with 24 metabolites measured on a daily basis in n = 4 independent industrial 2L fed batch cell culture processes for a therapeutic antibody production. This study is the first adaptation of a genome scale model for CHO cells to an industrial process, that successfully predicted cell phenotype. The tailored model predicted accurately both the exometabolomics data (r2 ≥ 0.8 for 96% of the considered metabolites) and growth rate (r2 = 0.91) of the industrial cell line. Flux distributions at different days of the process were analyzed for validation and suggestion of strategies for medium optimization. This study shows how to adapt a genome scale model to an industrial process and sheds light on the metabolic specificities of a high production process. The curated genome scale model is a great tool to gain insights into intracellular fluxes and to identify possible bottlenecks impacting cell performances during production process. The general use of genome scale models for modeling industrial recombinant cell lines is a long-term investment that will highly benefit process development and speed up time to market.


Assuntos
Técnicas de Cultura Celular por Lotes/métodos , Células CHO/metabolismo , Indústria Química , Genoma/genética , Aminoácidos/metabolismo , Animais , Simulação por Computador , Cricetinae , Cricetulus , Meios de Cultura , Metabolismo dos Lipídeos/genética , Engenharia Metabólica , Redes e Vias Metabólicas , Metabolômica , Modelos Biológicos , Modelos Teóricos , Fenótipo
8.
Metab Eng ; 52: 143-152, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30513349

RESUMO

Recombinant Chinese hamster ovary (CHO) cells are able to provide biopharmaceuticals that are essentially free of human viruses and have N-glycosylation profiles similar, but not identical, to humans. Due to differences in N-glycan moieties, two members of the serpin superfamily, alpha-1-antitrypsin (A1AT) and plasma protease C1 inhibitor (C1INH), are currently derived from human plasma for treating A1AT and C1INH deficiency. Deriving therapeutic proteins from human plasma is generally a cost-intensive process and also harbors a risk of transmitting infectious particles. Recombinantly produced A1AT and C1INH (rhA1AT, rhC1INH) decorated with humanized N-glycans are therefore of clinical and commercial interest. Here, we present engineered CHO cell lines producing rhA1AT or rhC1INH with fully humanized N-glycosylation profiles. This was achieved by combining CRISPR/Cas9-mediated disruption of 10 gene targets with overexpression of human ST6GAL1. We were able to show that the N-linked glyco-structures of rhA1AT and rhC1INH are homogeneous and similar to the structures obtained from plasma-derived A1AT and C1INH, marketed as Prolastin®-C and Cinryze®, respectively. rhA1AT and rhC1INH produced in our glyco-engineered cell line showed no detectable differences to their plasma-purified counterparts on SDS-PAGE and had similar enzymatic in vitro activity. The work presented here shows the potential of expanding the glyco-engineering toolbox for CHO cells to produce a wider variety of glycoproteins with fully humanized N-glycan profiles. We envision replacing plasma-derived A1AT and C1INH with recombinant versions and thereby decreasing our dependence on human donor blood, a limited and possibly unsafe protein source for patients.


Assuntos
Células CHO/metabolismo , Proteína Inibidora do Complemento C1/biossíntese , Engenharia Metabólica/métodos , alfa 1-Antitripsina/biossíntese , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Sistemas CRISPR-Cas , Cricetinae , Cricetulus , Glicosilação , Humanos , Proteínas Recombinantes/biossíntese , Sialiltransferases/biossíntese , Sialiltransferases/genética
9.
Sci Rep ; 8(1): 17957, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30560908

RESUMO

The increased interest in secondary metabolites (SMs) has driven a number of genome sequencing projects to elucidate their biosynthetic pathways. As a result, studies revealed that the number of secondary metabolite gene clusters (SMGCs) greatly outnumbers detected compounds, challenging current methods to dereplicate and categorize this amount of gene clusters on a larger scale. Here, we present an automated workflow for the genetic dereplication and analysis of secondary metabolism genes in fungi. Focusing on the secondary metabolite rich genus Aspergillus, we categorize SMGCs across genomes into SMGC families using network analysis. Our method elucidates the diversity and dynamics of secondary metabolism in section Nigri, showing that SMGC diversity within the section has the same magnitude as within the genus. Using our genome analysis we were able to predict the gene cluster responsible for biosynthesis of malformin, a potentiator of anti-cancer drugs, in 18 strains. To proof the general validity of our predictions, we developed genetic engineering tools in Aspergillus brasiliensis and subsequently verified the genes for biosynthesis of malformin.


Assuntos
Vias Biossintéticas , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Família Multigênica , Metabolismo Secundário/genética , Aspergillus/genética , Aspergillus/metabolismo , Análise por Conglomerados , Biologia Computacional/métodos , Mineração de Dados , Perfilação da Expressão Gênica , Engenharia Genética , Genômica/métodos , Anotação de Sequência Molecular
10.
ACS Synth Biol ; 7(9): 2148-2159, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30060646

RESUMO

Mammalian cells are widely used to express genes for basic biology studies and biopharmaceuticals. Current methods for generation of engineered cell lines introduce high genomic and phenotypic diversity, which hamper studies of gene functions and discovery of novel cellular mechanisms. Here, we minimized clonal variation by integrating a landing pad for recombinase-mediated cassette exchange site-specifically into the genome of CHO cells using CRISPR and generated subclones expressing four different recombinant proteins. The subclones showed low clonal variation with high consistency in growth, transgene transcript levels and global transcriptional response to recombinant protein expression, enabling improved studies of the impact of transgenes on the host transcriptome. Little variation over time in subclone phenotypes and transcriptomes was observed when controlling environmental culture conditions. The platform enables robust comparative studies of genome engineered CHO cell lines and can be applied to other mammalian cells for diverse biological, biomedical and biotechnological applications.


Assuntos
Engenharia Celular , Proteínas Recombinantes/metabolismo , Biologia de Sistemas/métodos , Animais , Células CHO , Sistemas CRISPR-Cas/genética , Cricetinae , Cricetulus , Eritropoetina/genética , Eritropoetina/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Proteínas Recombinantes/genética , Transcrição Gênica , Transcriptoma
11.
Biotechnol J ; 13(10): e1800111, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29862652

RESUMO

In production of recombinant proteins for biopharmaceuticals, N-glycosylation is often important for protein efficacy and patient safety. IgG with agalactosylated (G0)-N-glycans can improve the activation of the lectin-binding complement system and be advantageous in the therapy of lupus and virus diseases. In this study, the authors aimed to engineer CHO-S cells for the production of proteins with G0-N-glycans by targeting B4Gal-T isoform genes with CRISPR/Cas9. Indel mutations in genes encoding B4Gal-T1, -T2, and -T3 with and without a disrupted B4Gal-T4 sequence resulted in only ≈1% galactosylated N-glycans on total secreted proteins of 3-4 clones per genotype. The authors revealed that B4Gal-T4 is not active in N-glycan galactosylation in CHO-S cells. In the triple-KO clones, transiently expressed erythropoietin (EPO) and rituximab harbored only ≈6% and ≈3% galactosylated N-glycans, respectively. However, simultaneous disruption of B4Gal-T1 and -T3 may decrease cell growth. Altogether, the authors present the advantage of analyzing total secreted protein N-glycans after disrupting galactosyltransferases, followed by expressing recombinant proteins in selected clones with desired N-glycan profiles at a later stage. Furthermore, the authors provide a cell platform that prevalently glycosylates proteins with G0-N-glycans to further study the impact of agalactosylation on different in vitro and in vivo functions of recombinant proteins.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Polissacarídeos , Proteínas Recombinantes , Animais , Células CHO , Cricetulus , Expressão Gênica , Glicosilação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
12.
Biotechnol J ; 13(3): e1700499, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29393587

RESUMO

For over three decades, Chinese hamster ovary (CHO) cells have been the chosen expression platform for the production of therapeutic proteins with complex post-translational modifications. However, the metabolism of these cells is far from perfect and optimized, and requires substantial know how and process optimization and monitoring to perform efficiently. One of the main reasons for this is the production and accumulation of toxic and growth-inhibiting metabolites during culture. Lactate and ammonium are the most known, but many more have been identified. In this review, an overview of metabolites that deplete and accumulate throughout the course of cultivations with toxic and growth inhibitory effects to the cells is presented. Further, an overview of the CHO metabolism with emphasis to metabolic pathways of amino acids, glutathione (GSH), and related compounds which have growth-inhibiting and/or toxic effect on the cells is provided. Additionally, relevant publications which describe the applications of metabolomics as a powerful tool for revealing which reactions occur in the cell under certain conditions are surveyed and growth-inhibiting and toxic metabolites are identified. Also, a number of resources that describe the cellular mechanisms of CHO and are available on-line are presented. Finally, the application of this knowledge for bioprocess and medium development and cell line engineering is discussed.


Assuntos
Células CHO/metabolismo , Técnicas de Cultura de Células/métodos , Metabolômica , Proteínas Recombinantes/biossíntese , Aminoácidos/metabolismo , Animais , Cricetinae , Cricetulus , Alimentos , Ácido Láctico/metabolismo , Redes e Vias Metabólicas/genética , Processamento de Proteína Pós-Traducional/genética , Proteínas Recombinantes/genética
13.
Biotechnol J ; 13(3): e1700216, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29359860

RESUMO

The selection of clonally derived Chinese hamster ovary (CHO) cell lines with the highest production rate of recombinant glycoproteins remains a big challenge during early stages of cell line development. Different strategies using either product titer or product titer normalized to cell number are being used to assess suspension-adapted clones when grown statically in microtiter plates. However, no reported study so far has performed a direct head-to-head comparison of these two early reporters for predicting clone performance. Therefore, a screening platform for high-throughput analysis of titer and confluence of etanercept-producing clones is developed. Then an unbiased comparison of clone ranking based on either titer or titer normalized to confluence (TTC) is performed. Using two different suspension cultivation vessels, the authors demonstrate that titer- or TTC-based ranking gives rise to the selection of clones with similar volumetric productivity in batch cultures. Therefore, using both titer- and TTC-based ranking is proposed, allowing for selection of distinct clones with both high integral of viable cell density (IVCD) and high specific productivity features, respectively. This contributes to selection of a versatile panel of clones that can be further characterized and from which the final producer clone can be selected that best fits the production requirements.


Assuntos
Técnicas de Cultura Celular por Lotes/métodos , Etanercepte/metabolismo , Glicoproteínas/biossíntese , Proteínas Recombinantes/biossíntese , Animais , Células CHO , Contagem de Células , Cricetinae , Cricetulus , Etanercepte/química , Glicoproteínas/genética , Proteínas Recombinantes/genética
14.
Methods Mol Biol ; 1674: 147-161, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28921435

RESUMO

Fed-batch culture is the most commonly used upstream process in industry today for recombinant monoclonal antibody production using Chinese hamster ovary (CHO) cells. Developing and optimizing this process in the lab is crucial for establishing process knowledge, which enables rapid and predictable tech-transfer to manufacturing scale. In this chapter, we describe stepwise how to carry out fed-batch CHO cell culture for lab-scale antibody production.


Assuntos
Anticorpos Monoclonais/metabolismo , Proteínas Recombinantes/metabolismo , Animais , Formação de Anticorpos/fisiologia , Técnicas de Cultura Celular por Lotes/métodos , Reatores Biológicos , Células CHO , Técnicas de Cultura de Células/métodos , Linhagem Celular , Cricetulus
15.
Methods Mol Biol ; 1603: 209-226, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28493133

RESUMO

Chinese hamster ovary (CHO) cells have become the primary expression system for the production of complex recombinant proteins due to their long-term success in industrial scale production and generating appropriate protein N-glycans similar to that of humans. Control and optimization of protein N-glycosylation is crucial, as the structure of N-glycans can largely influence both biological and physicochemical properties of recombinant proteins. Protein N-glycosylation in CHO cell culture can be controlled and tuned by engineering medium, feed, culture process, as well as genetic elements of the cell. In this chapter, we will focus on how to carry out experiments for N-glycosylation modulation through medium and feed optimization. The workflow and typical methods involved in the experiment process will be presented.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura/química , Polissacarídeos/química , Proteínas Recombinantes/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Glicosilação
16.
Cell Syst ; 4(3): 262-275, 2017 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-28334575

RESUMO

Rational approaches to modifying cells to make molecules of interest are of substantial economic and scientific interest. Most of these efforts aim at the production of native metabolites, expression of heterologous biosynthetic pathways, or protein expression. Reviews of these topics have largely focused on individual strategies or cell types, but collectively they fall under the broad umbrella of a growing field known as cell factory engineering. Here we condense >130 reviews and key studies in the art into a meta-review of cell factory engineering. We identified 33 generic strategies in the field, all applicable to multiple types of cells and products, and proven successful in multiple major cell types. These apply to three major categories: production of native metabolites and/or bioactives, heterologous expression of biosynthetic pathways, and protein expression. This meta-review provides general strategy guides for the broad range of applications of rational engineering of cell factories.


Assuntos
Engenharia Celular/métodos , Engenharia Genética/métodos , Engenharia de Proteínas/métodos , Animais , Vias Biossintéticas/genética , Vias Biossintéticas/fisiologia , Humanos , Engenharia Metabólica/métodos
17.
BMC Syst Biol ; 11(1): 37, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28298216

RESUMO

BACKGROUND: Protein secretion is one of the most important processes in eukaryotes. It is based on a highly complex machinery involving numerous proteins in several cellular compartments. The elucidation of the cell biology of the secretory machinery is of great importance, as it drives protein expression for biopharmaceutical industry, a 140 billion USD global market. However, the complexity of secretory process is difficult to describe using a simple reductionist approach, and therefore a promising avenue is to employ the tools of systems biology. RESULTS: On the basis of manual curation of the literature on the yeast, human, and mouse secretory pathway, we have compiled a comprehensive catalogue of characterized proteins with functional annotation and their interconnectivity. Thus we have established the most elaborate reconstruction (RECON) of the functional secretion pathway network to date, counting 801 different components in mouse. By employing our mouse RECON to the CHO-K1 genome in a comparative genomic approach, we could reconstruct the protein secretory pathway of CHO cells counting 764 CHO components. This RECON furthermore facilitated the development of three alternative methods to study protein secretion through graphical visualizations of omics data. We have demonstrated the use of these methods to identify potential new and known targets for engineering improved growth and IgG production, as well as the general observation that CHO cells seem to have less strict transcriptional regulation of protein secretion than healthy mouse cells. CONCLUSIONS: The RECON of the secretory pathway represents a strong tool for interpretation of data related to protein secretion as illustrated with transcriptomic data of Chinese Hamster Ovary (CHO) cells, the main platform for mammalian protein production.


Assuntos
Biologia Computacional/métodos , Perfilação da Expressão Gênica , Via Secretória/genética , Animais , Células CHO , Cricetinae , Cricetulus , Ontologia Genética , Camundongos
18.
Sci Rep ; 7: 40388, 2017 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-28091612

RESUMO

Recombinant protein production coopts the host cell machinery to provide high protein yields of industrial enzymes or biotherapeutics. However, since protein translation is energetically expensive and tightly controlled, it is unclear if highly expressed recombinant genes are translated as efficiently as host genes. Furthermore, it is unclear how the high expression impacts global translation. Here, we present the first genome-wide view of protein translation in an IgG-producing CHO cell line, measured with ribosome profiling. Through this we found that our recombinant mRNAs were translated as efficiently as the host cell transcriptome, and sequestered up to 15% of the total ribosome occupancy. During cell culture, changes in recombinant mRNA translation were consistent with changes in transcription, demonstrating that transcript levels influence specific productivity. Using this information, we identified the unnecessary resistance marker NeoR to be a highly transcribed and translated gene. Through siRNA knock-down of NeoR, we improved the production- and growth capacity of the host cell. Thus, ribosomal profiling provides valuable insights into translation in CHO cells and can guide efforts to enhance protein production.


Assuntos
Proteínas/metabolismo , Ribossomos/metabolismo , Animais , Células CHO , Contagem de Células , Proliferação de Células/genética , Sobrevivência Celular/genética , Cricetinae , Cricetulus , Técnicas de Silenciamento de Genes , Imunoglobulina G/metabolismo , Nucleotídeos/metabolismo , Biossíntese de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Transcrição Gênica
19.
Toxicon ; 126: 79-89, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28017694

RESUMO

Antivenoms against bites and stings from snakes, spiders, and scorpions are associated with immunological side effects and high cost of production, since these therapies are still derived from the serum of hyper-immunized production animals. Biotechnological innovations within envenoming therapies are thus warranted, and phage display technology may be a promising avenue for bringing antivenoms into the modern era of biologics. Although phage display technology represents a robust and high-throughput approach for the discovery of antibody-based antitoxins, several pitfalls may present themselves when animal toxins are used as targets for phage display selection. Here, we report selected critical challenges from our own phage display experiments associated with biotinylation of antigens, clone picking, and the presence of amber codons within antibody fragment structures in some phage display libraries. These challenges may be detrimental to the outcome of phage display experiments, and we aim to help other researchers avoiding these pitfalls by presenting their solutions.


Assuntos
Antivenenos/química , Técnicas de Visualização da Superfície Celular/métodos , Venenos de Serpentes/química , Biotinilação , Fragmentos de Imunoglobulinas/química , Modelos Teóricos
20.
PLoS One ; 11(8): e0161199, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27551732

RESUMO

Polyketide synthases (PKSs) and nonribosomal peptide synthetases (NRPSs) each give rise to a vast array of complex bioactive molecules with further complexity added by the existence of natural PKS-NRPS fusions. Rational genetic engineering for the production of natural product derivatives is desirable for the purpose of incorporating new functionalities into pre-existing molecules, or for optimization of known bioactivities. We sought to expand the range of natural product diversity by combining modules of PKS-NRPS hybrids from different hosts, hereby producing novel synthetic natural products. We succeeded in the construction of a functional cross-species chimeric PKS-NRPS expressed in Aspergillus nidulans. Module swapping of the two PKS-NRPS natural hybrids CcsA from Aspergillus clavatus involved in the biosynthesis of cytochalasin E and related Syn2 from rice plant pathogen Magnaporthe oryzae lead to production of novel hybrid products, demonstrating that the rational re-design of these fungal natural product enzymes is feasible. We also report the structure of four novel pseudo pre-cytochalasin intermediates, niduclavin and niduporthin along with the chimeric compounds niduchimaeralin A and B, all indicating that PKS-NRPS activity alone is insufficient for proper assembly of the cytochalasin core structure. Future success in the field of biocombinatorial synthesis of hybrid polyketide-nonribosomal peptides relies on the understanding of the fundamental mechanisms of inter-modular polyketide chain transfer. Therefore, we expressed several PKS-NRPS linker-modified variants. Intriguingly, the linker anatomy is less complex than expected, as these variants displayed great tolerance with regards to content and length, showing a hitherto unreported flexibility in PKS-NRPS hybrids, with great potential for synthetic biology-driven biocombinatorial chemistry.


Assuntos
Aspergillus nidulans/genética , Engenharia Genética , Peptídeo Sintases/genética , Policetídeo Sintases/genética , Aspergillus nidulans/enzimologia , Produtos Biológicos , Citocalasinas/biossíntese , Regulação Enzimológica da Expressão Gênica , Genes Sintéticos/genética , Magnaporthe/enzimologia , Magnaporthe/genética , Peptídeo Sintases/biossíntese , Policetídeo Sintases/biossíntese , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA