Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Biotechnol Bioeng ; 120(7): 1746-1761, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36987713

RESUMO

Protein expression from stably transfected Chinese hamster ovary (CHO) clones is an established but time-consuming method for manufacturing therapeutic recombinant proteins. The use of faster, alternative approaches, such as non-clonal stable pools, has been restricted due to lower productivity and longstanding regulatory guidelines. Recently, the performance of stable pools has improved dramatically, making them a viable option for quickly producing drug substance for GLP-toxicology and early-phase clinical trials in scenarios such as pandemics that demand rapid production timelines. Compared to stable CHO clones which can take several months to generate and characterize, stable pool development can be completed in only a few weeks. Here, we compared the productivity and product quality of trimeric SARS-CoV-2 spike protein ectodomains produced from stable CHO pools or clones. Using a set of biophysical and biochemical assays we show that product quality is very similar and that CHO pools demonstrate sufficient productivity to generate vaccine candidates for early clinical trials. Based on these data, we propose that regulatory guidelines should be updated to permit production of early clinical trial material from CHO pools to enable more rapid and cost-effective clinical evaluation of potentially life-saving vaccines.


Assuntos
COVID-19 , SARS-CoV-2 , Cricetinae , Animais , Humanos , Cricetulus , SARS-CoV-2/metabolismo , Células CHO , Anticorpos Monoclonais , Vacinas contra COVID-19/genética , COVID-19/prevenção & controle , Proteínas Recombinantes/metabolismo , Vacinas de Subunidades Antigênicas/genética
2.
Biotechnol Bioeng ; 119(8): 2206-2220, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35509261

RESUMO

Some effector functions prompted by immunoglobulin G (IgG) antibodies, such as antibody-dependent cell-mediated cytotoxicity (ADCC), strongly depend on the N-glycans linked to asparagine 297 of the Fc region of the protein. A single α-(1,6)-fucosyltransferase (FUT8) is responsible for catalyzing the addition of an α-1,6-linked fucose residue to the first GlcNAc residue of the N-linked glycans. Antibodies missing this core fucose show a significantly enhanced ADCC and increased antitumor activity, which could help reduce therapeutic dose requirement, potentially translating into reduced safety concerns and manufacturing costs. Several approaches have been developed to modify glycans and improve the biological functions of antibodies. Here, we demonstrate that expression of a membrane-associated anti-FUT8 intrabody engineered to reside in the endoplasmic reticulum and Golgi apparatus can efficiently reduce FUT8 activity and therefore the core-fucosylation of the Fc N-glycan of an antibody. IgG1-producing CHO cells expressing the intrabody secrete antibodies with reduced core fucosylation as demonstrated by lectin blot analysis and UPLC-HILIC glycan analysis. Cells engineered to inhibit directly and specifically alpha-(1,6)-fucosyltransferase activity allows for the production of g/L levels of IgGs with strongly enhanced ADCC effector function, for which the level of fucosylation can be selected. The quick and efficient method described here should have broad practical applicability for the development of next-generation therapeutic antibodies with enhanced effector functions.


Assuntos
Fucose , Fucosiltransferases , Animais , Anticorpos Monoclonais/química , Células CHO , Cricetinae , Cricetulus , Fucose/metabolismo , Fucosiltransferases/genética , Imunoglobulina G/química , Polissacarídeos
3.
Methods Mol Biol ; 2810: 99-121, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38926275

RESUMO

The continuous improvement of expression platforms is necessary to respond to the increasing demand for recombinant proteins that are required to carry out structural or functional studies as well as for their characterization as biotherapeutics. While transient gene expression (TGE) in mammalian cells constitutes a rapid and well-established approach, non-clonal stably transfected cells, or "pools," represent another option, which is especially attractive when recurring productions of the same protein are required. From a culture volume of just a few liters, stable pools can provide hundreds of milligrams to gram quantities of high-quality secreted recombinant proteins.In this chapter, we describe a highly efficient and cost-effective procedure for the generation of Chinese Hamster Ovary cell stable pools expressing secreted recombinant proteins using commercially available serum-free media and polyethylenimine (PEI) as the transfection reagent. As a specific example of how this protocol can be applied, the production and downstream purification of recombinant His-tagged trimeric SARS-CoV-2 spike protein ectodomain (SmT1) are described.


Assuntos
Cricetulus , Polietilenoimina , Proteínas Recombinantes , Glicoproteína da Espícula de Coronavírus , Transfecção , Células CHO , Animais , Proteínas Recombinantes/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Transfecção/métodos , Polietilenoimina/química , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/biossíntese , Glicoproteína da Espícula de Coronavírus/isolamento & purificação , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Cricetinae , Meios de Cultura Livres de Soro
4.
Commun Chem ; 6(1): 189, 2023 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-37684364

RESUMO

Glycosylation is a key quality attribute that must be closely monitored for protein therapeutics. Established assays such as HILIC-Fld of released glycans and LC-MS of glycopeptides work well for glycoproteins with a few glycosylation sites but are less amenable for those with multiple glycosylation sites, resulting in complex datasets that are time consuming to generate and difficult to analyze. As part of efforts to improve preparedness for future pandemics, researchers are currently assessing where time can be saved in the vaccine development and production process. In this context, we evaluated if neutral and acidic monosaccharides analysis via HPAEC-PAD could be used as a rapid and robust alternative to LC-MS and HILIC-Fld for monitoring glycosylation between protein production batches. Using glycoengineered spike proteins we show that the HPAEC-PAD monosaccharide assays could quickly and reproducibly detect both major and minor glycosylation differences between batches. Moreover, the monosaccharide results aligned well with those obtained by HILIC-Fld and LC-MS.

5.
Methods ; 55(1): 44-51, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21539918

RESUMO

Transient gene expression in mammalian cells is a valuable alternative to stable cell lines for the rapid production of large amounts of recombinant proteins. While the establishment of stable cell lines takes 2-6 months, milligram amounts of protein can be obtained within a week following transfection. The polycation polyethylenimine (PEI) is one of the most utilized reagents for small- to large-scale transfections as it is simple to use and, when combined with optimized expression vectors and cell lines, provides high transfection efficiency and titers. As with most transfection reagents, PEI-mediated transfection involves the formation of nanoparticles (polyplexes) which are obtained by its mixing with plasmid DNA. A short incubation period that allows polyplexes to reach their optimal size is performed prior to their addition to the culture. As the quality of polyplexes directly impacts transfection efficiency and productivity, their formation complicates scalability and automation of the process, especially when performed in large-scale bioreactors or small-scale high-throughput formats. To avoid variations in transfection efficiency and productivity that arise from polyplexes formation step, we have optimized the conditions for their creation directly in the culture by the consecutive addition of DNA and PEI. This simplified approach is directly transferable from suspension cultures grown in 6-well plates to shaker flasks and 5-L WAVE bioreactors. As it minimizes the number of steps and does not require an incubation period for polyplex formation, it is also suitable for automation using static cultures in 96-well plates. This "direct" transfection method thus provides a robust platform for both high-throughput expression and large-scale production of recombinant proteins.


Assuntos
Técnicas de Cultura de Células/métodos , Ensaios de Triagem em Larga Escala , Polietilenoimina/química , Proteínas Recombinantes/biossíntese , Transfecção/métodos , Animais , Reatores Biológicos , Linhagem Celular , DNA/química , DNA/genética , Expressão Gênica , Vetores Genéticos/química , Humanos , Mamíferos , Nanopartículas/química , Plasmídeos/química , Plasmídeos/genética , Polietilenoimina/metabolismo , Proteínas Recombinantes/genética
6.
Commun Biol ; 5(1): 933, 2022 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-36085335

RESUMO

Nanobodies offer several potential advantages over mAbs for the control of SARS-CoV-2. Their ability to access cryptic epitopes conserved across SARS-CoV-2 variants of concern (VoCs) and feasibility to engineer modular, multimeric designs, make these antibody fragments ideal candidates for developing broad-spectrum therapeutics against current and continually emerging SARS-CoV-2 VoCs. Here we describe a diverse collection of 37 anti-SARS-CoV-2 spike glycoprotein nanobodies extensively characterized as both monovalent and IgG Fc-fused bivalent modalities. The nanobodies were collectively shown to have high intrinsic affinity; high thermal, thermodynamic and aerosolization stability; broad subunit/domain specificity and cross-reactivity across existing VoCs; wide-ranging epitopic and mechanistic diversity and high and broad in vitro neutralization potencies. A select set of Fc-fused nanobodies showed high neutralization efficacies in hamster models of SARS-CoV-2 infection, reducing viral burden by up to six orders of magnitude to below detectable levels. In vivo protection was demonstrated with anti-RBD and previously unreported anti-NTD and anti-S2 nanobodies. This collection of nanobodies provides a potential therapeutic toolbox from which various cocktails or multi-paratopic formats could be built to combat multiple SARS-CoV-2 variants.


Assuntos
COVID-19 , Anticorpos de Domínio Único , Animais , Anticorpos Monoclonais , Cricetinae , Humanos , SARS-CoV-2/genética , Anticorpos de Domínio Único/genética
7.
J Biotechnol ; 326: 21-27, 2021 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-33301853

RESUMO

Recombinant forms of the spike protein of SARS-CoV-2 and related viruses have proven difficult to produce with good yields in mammalian cells. Given the panoply of potential COVID-19 diagnostic tools and therapeutic candidates that require purified spike protein and its importance for ongoing SARS-CoV-2 research, we have explored new approaches for spike production and purification. Three transient gene expression methods based on PEI-mediated transfection of CHO or HEK293 cells in suspension culture in chemically-defined media were compared for rapid production of full-length SARS-CoV-2 spike ectodomain. A high-cell-density protocol using DXB11-derived CHOBRI/55E1 cells gave substantially better yields than the other methods. Different forms of the spike ectodomain were expressed, including the wild-type SARS-CoV-2 sequence and a mutated form (to favor expression of the full-length spike ectodomain stabilized in pre-fusion conformation), with and without fusion to putative trimerization domains. An efficient two-step affinity purification method was also developed. Ultimately, we have been able to produce highly homogenous preparations of full-length spike, both monomeric and trimeric, with yields of 100-150 mg/L in the harvested medium. The speed and productivity of this method support further development of CHO-based approaches for recombinant spike protein manufacturing.


Assuntos
Domínios Proteicos , Proteínas Recombinantes , Glicoproteína da Espícula de Coronavírus/genética , Animais , Células CHO , Cricetulus , Expressão Gênica , Células HEK293 , Humanos , SARS-CoV-2 , Transfecção
8.
BMC Biotechnol ; 8: 65, 2008 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-18752669

RESUMO

BACKGROUND: Mammalian cells are becoming the prevailing expression system for the production of recombinant proteins because of their capacity for proper protein folding, assembly, and post-translational modifications. These systems currently allow high volumetric production of monoclonal recombinant antibodies in the range of grams per litre. However their use for large-scale expression of cytokines typically results in much lower volumetric productivity. RESULTS: We have engineered a HEK293 cell clone for high level production of human recombinant glycosylated IFNalpha2b and developed a rapid and efficient method for its purification. This clone steadily produces more than 200 mg (up to 333 mg) of human recombinant IFNalpha2b per liter of serum-free culture, which can be purified by a single-step cation-exchange chromatography following media acidification and clarification. This rapid procedure yields 98% pure IFNalpha2b with a recovery greater than 70%. Purified IFNalpha2b migrates on SDS-PAGE as two species, a major 21 kDa band and a minor 19 kDa band. N-terminal sequences of both forms are identical and correspond to the expected mature protein. Purified IFNalpha2b elutes at neutral pH as a single peak with an apparent molecular weight of 44,000 Da as determined by size-exclusion chromatography. The presence of intramolecular and absence of intermolecular disulfide bridges is evidenced by the fact that non-reduced IFNalpha2b has a greater electrophoretic mobility than the reduced form. Treatment of purified IFNalpha2b with neuraminidase followed by O-glycosidase both increases electrophoretic mobility, indicating the presence of sialylated O-linked glycan. A detailed analysis of glycosylation by mass spectroscopy identifies disialylated and monosialylated forms as the major constituents of purified IFNalpha2b. Electron transfer dissociation (ETD) shows that the glycans are linked to the expected threonine at position 106. Other minor glycosylated forms and non-sialylated species are also detected, similar to IFNalpha2b produced naturally by lymphocytes. Further, the HEK293-produced IFNalpha2b is biologically active as shown with reporter gene and antiviral assays. CONCLUSION: These results show that the HEK293 cell line is an efficient and valuable host for the production of biologically active and glycosylated human IFNalpha2b.


Assuntos
Clonagem Molecular/métodos , Interferon-alfa/metabolismo , Rim/fisiologia , Engenharia de Proteínas/métodos , Proteínas Recombinantes/metabolismo , Linhagem Celular , Humanos , Interferon alfa-2 , Interferon-alfa/genética
9.
J Virol Methods ; 148(1-2): 106-14, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18079009

RESUMO

Helper-dependent adenovirus (HDAd), deleted in all viral protein-coding sequences has been designed to reduce immune response and favor long-term expression of therapeutic genes in clinical programs. Its production requires co-infection of E1-complementing cells with helper adenovirus (HAd). Significant progresses have been made in the molecular design of HDAd, but large scale production remains a challenge. In this work, a scalable system for HDAd production is designed and evaluated focusing on the co-infection step. A human embryo kidney 293 (293) derived cell line, the 293SF/FLPe was generated to produce efficiently HDAd while restricting the packaging of HAd. This cell line was adapted to grow in suspension and in serum-free medium. Multiplicity of infection (MOI) of HDAd ranging from 0.1 to 50 was evaluated in presence of HAd at a MOI of 5. Optimal MOIs for HDAd amplification were found in the range of 5-10. HAd contamination was only 1%. These results were validated in a 3 L bioreactor under controlled operating conditions where a higher HDAd yield of 2.6 x 10(9) viral particles (VP)/mL or 3.5 x 10(8) infectious units (IU)/mL of HDAd was obtained.


Assuntos
Adenoviridae/crescimento & desenvolvimento , Cultura de Vírus/métodos , Proteínas E1 de Adenovirus/genética , Contagem de Células , Linhagem Celular , Sobrevivência Celular , Meios de Cultura Livres de Soro , Vetores Genéticos , Vírus Auxiliares/fisiologia , Humanos , Transdução Genética
10.
J Biotechnol ; 281: 39-47, 2018 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-29886030

RESUMO

For pre-clinical evaluation of biotherapeutic candidates, protein production by transient gene expression (TGE) in Chinese Hamster Ovary (CHO) cells offers important advantages, including the capability of rapidly and cost-effectively generating recombinant proteins that are highly similar to those produced in stable CHO clones. We have established a novel CHO clone (CHO-3E7) expressing a form of the Epstein-Barr virus nuclear antigen-1 (EBNA-1) with improved TGE productivity relative to parental CHO cells. Taking advantage of a new transfection-compatible media formulation that permits prolonged, high-density culture, we optimized transfection parameters (cell density, plasmid vector and polyethylenimine concentrations) and post-transfection culture conditions to establish a new, high-performing process for rapid protein production. The growth media is chemically defined, and a single hydrolysate feed is added post-transfection, followed by periodic glucose supplementation. This method gave significantly higher yields than our standard low-cell density, F17-based CHO-3E7 TGE method, averaging several hundred mg/l for a panel of recombinant proteins and antibodies. Purified antibodies produced using the two methods had distinct glycosylation profiles but showed identical target binding kinetics by SPR. Key advantages of this new protein production platform include the cost-effectiveness of the transfection reagent, the commercial availability of the culture media and the ability to perform high-cell-density transfection without media change.


Assuntos
Antígenos Nucleares do Vírus Epstein-Barr/genética , Polietilenoimina , Transfecção/métodos , Trastuzumab/biossíntese , Animais , Células CHO , Contagem de Células , Cricetulus , Expressão Gênica
11.
J Biotechnol ; 255: 16-27, 2017 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-28625678

RESUMO

To rapidly produce large amounts of recombinant proteins, the generation of stable Chinese Hamster Ovary (CHO) cell pools represents a useful alternative to large-scale transient gene expression (TGE). We have developed a cell line (CHOBRI/rcTA) allowing the inducible expression of recombinant proteins, based on the cumate gene switch. After the identification of optimal plasmid DNA topology (supercoiled vs linearized plasmid) for PEIpro™ mediated transfection and of optimal conditions for methionine sulfoximine (MSX) selection, we were able to generate CHOBRI/rcTA pools producing high levels of recombinant proteins. Volumetric productivities of up to 900mg/L were reproducibly achieved for a Fc fusion protein and up to 350mg/L for an antibody after 14days post-induction in non-optimized fed-batch cultures. In addition, we show that CHO pool volumetric productivities are not affected by a freeze-thaw cycle or following maintenance in culture for over one month in the presence of MSX. Finally, we demonstrate that volumetric protein production with the CR5 cumate-inducible promoter is three- to four-fold higher than with the human CMV or hybrid EF1α-HTLV constitutive promoters. These results suggest that the cumate-inducible CHOBRI/rcTA stable pool platform is a powerful and robust system for the rapid production of gram amounts of recombinant proteins.


Assuntos
Plasmídeos/genética , Engenharia de Proteínas/métodos , Proteínas Recombinantes/metabolismo , Animais , Técnicas de Cultura Celular por Lotes , Células CHO , Cricetinae , Cricetulus , Expressão Gênica , Vetores Genéticos , Metionina Sulfoximina/farmacologia , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética
12.
Nucleic Acids Res ; 30(2): E9, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11788735

RESUMO

A scalable transfection procedure using polyethylenimine (PEI) is described for the human embryonic kidney 293 cell line grown in suspension. Green fluorescent protein (GFP) and human placental secreted alkaline phosphatase (SEAP) were used as reporter genes to monitor transfection efficiency and productivity. Up to 75% of GFP-positive cells were obtained using linear or branched 25 kDa PEI. The 293 cell line and two genetic variants, either expressing the SV40 large T-antigen (293T) or the Epstein-Barr virus (EBV) EBNA1 protein (293E), were tested for protein expression. The highest expression level was obtained with 293E cells using the EBV oriP-containing plasmid pCEP4. We designed the pTT vector, an oriP-based vector having an improved cytomegalovirus expression cassette. Using this vector, 10- and 3-fold increases in SEAP expression was obtained in 293E cells compared with pcDNA3.1 and pCEP4 vectors, respectively. The presence of serum had a positive effect on gene transfer and expression. Transfection of suspension-growing cells was more efficient with linear PEI and was not affected by the presence of medium conditioned for 24 h. Using the pTT vector, >20 mg/l of purified His-tagged SEAP was recovered from a 3.5 l bioreactor. Intracellular proteins were also produced at levels as high as 50 mg/l, representing up to 20% of total cell proteins.


Assuntos
Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Expressão Gênica , Proteínas Recombinantes/biossíntese , Fosfatase Alcalina/biossíntese , Fosfatase Alcalina/genética , Fosfatase Alcalina/isolamento & purificação , Fosfatase Alcalina/metabolismo , Animais , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/metabolismo , Reatores Biológicos , Biotecnologia/métodos , Western Blotting , Adesão Celular , Contagem de Células , Linhagem Celular Transformada , Cromatografia de Afinidade , Meios de Cultivo Condicionados/metabolismo , Citomegalovirus/genética , Antígenos Nucleares do Vírus Epstein-Barr/genética , Citometria de Fluxo , Proteínas Fúngicas/análise , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/genética , Genes Reporter/genética , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Camundongos , Peso Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Transfecção , Proteínas Virais/análise , Proteínas Virais/biossíntese , Proteínas Virais/genética
13.
PLoS One ; 8(8): e72800, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977353

RESUMO

Foot-and-mouth disease (FMD) is a highly contagious disease of cloven-hoofed animals. It produces severe economic losses in the livestock industry. Currently available vaccines are based on inactivated FMD virus (FMDV). The use of empty capsids as a subunit vaccine has been reported to be a promising candidate because it avoids the use of virus in the vaccine production and conserves the conformational epitopes of the virus. In this report, we explored transient gene expression (TGE) in serum-free suspension-growing mammalian cells for the production of FMDV recombinant empty capsids as a subunit vaccine. The recombinant proteins produced, assembled into empty capsids and induced protective immune response against viral challenge in mice. Furthermore, they were recognized by anti-FMDV bovine sera. By using this technology, we were able to achieve expression levels that are compatible with the development of a vaccine. Thus, TGE of mammalian cells is an easy to perform, scalable and cost-effective technology for the production of a recombinant subunit vaccine against FMDV.


Assuntos
Capsídeo/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Vírus da Febre Aftosa/genética , Expressão Gênica/efeitos dos fármacos , Mamíferos/virologia , Animais , Antígenos Virais/imunologia , Western Blotting , Bovinos , Proliferação de Células , Febre Aftosa/imunologia , Febre Aftosa/prevenção & controle , Febre Aftosa/virologia , Vírus da Febre Aftosa/crescimento & desenvolvimento , Vírus da Febre Aftosa/imunologia , Vetores Genéticos , Genoma Viral/genética , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/metabolismo , Suspensões , Transfecção , Vacinação , Vírion/metabolismo
14.
Methods Mol Biol ; 911: 287-303, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22886259

RESUMO

Camelid single domain antibodies fused to noncamelid Fc regions, also called chimeric heavy chain antibodies (cHCAb), offer great potential as therapeutic and diagnostic candidates due to their relatively small size (80 kDa) and intact Fc. In this chapter, we describe two approaches, limiting dilution and minipools, for generating nonamplified Chinese hamster ovary cell lines stably expressing cHCAb in suspension and serum-free cultures using a stringent antibiotic selection. Neither of the protocols necessitates the acquisition or implementation of expensive automated infrastructures and thus could be applied in any lab with minimal cell culture setup. The given protocol allows the isolation of stable clones capable of generating up to 100 mg/L of antibody in batch mode performed in shaker flasks.


Assuntos
Anticorpos Monoclonais/genética , Cadeias Pesadas de Imunoglobulinas/genética , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/metabolismo , Células CHO , Cricetinae , Expressão Gênica , Ordem dos Genes , Vetores Genéticos/genética , Cadeias Pesadas de Imunoglobulinas/isolamento & purificação , Cadeias Pesadas de Imunoglobulinas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
15.
Biotechnol Bioeng ; 84(3): 332-42, 2003 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-12968287

RESUMO

Large-scale transient transfection of mammalian cells is a recent and powerful technology for the fast production of milligram amounts of recombinant proteins (r-proteins). As many r-proteins used for therapeutic and structural studies are naturally secreted or engineered to be secreted, a cost-effective serum-free culture medium that allows their efficient expression and purification is required. In an attempt to design such a serum-free medium, the effect of nine protein hydrolysates on cell proliferation, transfection efficiency, and volumetric productivity was evaluated using green fluorescent protein (GFP) and human placental secreted alkaline phosphate (SEAP) as reporter genes. The suspension growing, serum-free adapted HEK293SF-3F6 cell line was stably transfected with an EBNA1-expression vector to increase protein expression when using EBV oriP bearing plasmids. Compared to our standard serum-free medium, concomitant addition of the gelatin peptone N3 and removal of BSA slightly enhanced transfection efficiency and significantly increased volumetric productivity fourfold. Using the optimized medium formulation, transfection efficiencies between 40-60% were routinely obtained and SEAP production reached 18 mg/L(-1). To date, we have successfully produced and purified over fifteen r-proteins from 1-14-L bioreactors using this serum-free system. As examples, we describe the scale-up of two secreted his-tagged r-proteins Tie-2 and Neuropilin-1 extracellular domains (ED) in bioreactors. Each protein was successfully purified to >95% purity following a single immobilized metal affinity chromatography (IMAC) step. In contrast, purification of Tie-2 and Neuropilin-1 produced in serum-containing medium was much less efficient. Thus, the use of our new serum-free EBNA1 cell line with peptone-enriched serum-free medium significantly improves protein expression compared to peptone-less medium, and significantly increases their purification efficiency compared to serum-containing medium. This eliminates labor-intensive and expensive chromatographic steps, and allows for the simple, reliable, and extremely fast production of milligram amounts of r-proteins within 5 days posttransfection.


Assuntos
Reatores Biológicos , Técnicas de Cultura de Células/métodos , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Rim/crescimento & desenvolvimento , Rim/metabolismo , Peptonas/metabolismo , Engenharia de Proteínas/métodos , Transfecção/métodos , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Meios de Cultura , Meios de Cultura Livres de Soro/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/genética , Humanos , Rim/citologia , Rim/efeitos dos fármacos , Neuropilina-1/biossíntese , Neuropilina-1/genética , Peptonas/farmacologia , Receptor TIE-2/biossíntese , Receptor TIE-2/genética , Proteínas Recombinantes/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA