Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Microb Biotechnol ; 16(5): 893-900, 2023 05.
Article in English | MEDLINE | ID: mdl-35830603

ABSTRACT

Early-stage inclusion body formation is still mysterious. Literature is ambiguous about the existence of rod-shaped protein aggregates, a potential sponge-like inclusion body scaffold as well as the number of inclusion bodies per Escherichia coli cell. In this study, we verified the existence of rod-shaped inclusion bodies, confirmed their porous morphology, the presence of multiple protein aggregates per cell and modelled inclusion body formation as function of the number of generations.


Subject(s)
Escherichia coli , Protein Aggregates , Escherichia coli/genetics , Escherichia coli/metabolism , Inclusion Bodies/metabolism , Recombinant Proteins/metabolism
3.
Biotechnol Bioeng ; 118(10): 3847-3859, 2021 10.
Article in English | MEDLINE | ID: mdl-34129251

ABSTRACT

Predicting the fate of individual cells among a microbial population (i.e., growth and gene expression) remains a challenge, especially when this population is exposed to very dynamic environmental conditions, such as those encountered during continuous cultivation. Indeed, the dynamic nature of a continuous cultivation process implies the potential diversification of the microbial population resulting in genotypic and phenotypic heterogeneity. The present work focused on the induction of the arabinose operon in Escherichia coli as a model system to study this diversification process in continuous cultivations. As a preliminary step, the green fluorescent protein (GFP) level triggered by an arabinose-inducible ParaBAD promoter was tracked by flow cytometry in chemostat cultivations with glucose-arabinose co-feeding. For a wide range of glucose-arabinose co-feeding concentrations in the chemostats, the simultaneous occurrence of GFP positive and negative subpopulation was observed. In the second set of experiments, continuous cultivation was performed by adding glucose continuously and arabinose based on the capability of individual cells to switch from low GFP to high GFP expression states, performed with a technology setup called segregostat. In the segregostat cultivation mode, on-line flow cytometry analysis was used for adjusting the arabinose/glucose transitions based on the phenotypic switching profiles of the microbial population. This strategy allowed finding an appropriate arabinose pulsing frequency, leading to prolonged maintenance of the induction level with a limited increase in the phenotypic diversity for more than 60 generations. The results suggest that the steady forcing of individual cells into a given phenotypic trajectory may not be the best strategy for controlling cell populations. Instead, allowing individual cells to switch periodically around a predefined threshold seems to be a more robust strategy leading to oscillations, but within a predictable cell population behavior range.


Subject(s)
Escherichia coli K12 , Green Fluorescent Proteins/biosynthesis , Promoter Regions, Genetic , Arabinose/genetics , Arabinose/metabolism , Escherichia coli K12/genetics , Escherichia coli K12/growth & development , Green Fluorescent Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
4.
Sci Rep ; 11(1): 11477, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34075099

ABSTRACT

In many industrial sectors continuous processing is already the golden standard to maximize productivity. However, when working with living cells, subpopulation formation causes instabilities in long-term cultivations. In cascaded continuous cultivation, biomass formation and recombinant protein expression can be spatially separated. This cultivation mode was found to facilitate stable protein expression using microbial hosts, however mechanistic knowledge of this cultivation strategy is scarce. In this contribution we present a method workflow to reduce workload and accelerate the establishment of stable continuous processes with E. coli BL21(DE3) exclusively based on bioengineering methods.


Subject(s)
Biomass , Escherichia coli/growth & development , Bioengineering , Escherichia coli/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
5.
Front Bioeng Biotechnol ; 8: 573607, 2020.
Article in English | MEDLINE | ID: mdl-33240864

ABSTRACT

Recombinant protein production with Escherichia coli is usually carried out in fed-batch mode in industry. As set-up and cleaning of equipment are time- and cost-intensive, it would be economically and environmentally favorable to reduce the number of these procedures. Switching from fed-batch to continuous biomanufacturing with microbials is not yet applied as these cultivations still suffer from time-dependent variations in productivity. Repetitive fed-batch process technology facilitates critical equipment usage, reduces the environmental fingerprint and potentially increases the overall space-time yield. Surprisingly, studies on repetitive fed-batch processes for recombinant protein production can be found for yeasts only. Knowledge on repetitive fed-batch cultivation technology for recombinant protein production in E. coli is not available until now. In this study, a mixed feed approach, enabling repetitive fed-batch technology for recombinant protein production in E. coli, was developed. Effects of the cultivation mode on the space-time yield for a single-cycle fed-batch, a two-cycle repetitive fed-batch, a three-cycle repetitive fed batch and a chemostat cultivation were investigated. For that purpose, we used two different E. coli strains, expressing a model protein in the cytoplasm or in the periplasm, respectively. Our results demonstrate that a repetitive fed-batch for E. coli leads to a higher space-time yield compared to a single-cycle fed-batch and can potentially outperform continuous biomanufacturing. For the first time, we were able to show that repetitive fed-batch technology is highly suitable for recombinant protein production in E. coli using our mixed feeding approach, as it potentially (i) improves product throughput by using critical equipment to its full capacity and (ii) allows implementation of a more economic process by reducing cleaning and set-up times.

6.
Article in English | MEDLINE | ID: mdl-32903513

ABSTRACT

Continuous cultivation with Escherichia coli has several benefits compared to classical fed-batch cultivation. The economic benefits would be a stable process, which leads to time independent quality of the product, and hence ease the downstream process. However, continuous biomanufacturing with E. coli is known to exhibit a drop of productivity after about 4-5 days of cultivation depending on dilution rate. These cultivations are generally performed on glucose, being the favorite carbon source for E. coli and used in combination with isopropyl ß-D-1 thiogalactopyranoside (IPTG) for induction. In recent works, harsh induction with IPTG was changed to softer induction using lactose for T7-based plasmids, with the result of reducing the metabolic stress and tunability of productivity. These mixed feed systems based on glucose and lactose result in high amounts of correctly folded protein. In this study we used different mixed feed systems with glucose/lactose and glycerol/lactose to investigate productivity of E. coli based chemostats. We tested different strains producing three model proteins, with the final aim of a stable long-time protein expression. While glucose fed chemostats showed the well-known drop in productivity after a certain process time, glycerol fed cultivations recovered productivity after about 150 h of induction, which corresponds to around 30 generation times. We want to further highlight that the cellular response upon galactose utilization in E. coli BL21(DE3), might be causing fluctuating productivity, as galactose is referred to be a weak inducer. This "Lazarus" phenomenon has not been described in literature before and may enable a stabilization of continuous cultivation with E. coli using different carbon sources.

7.
J Pharm Biomed Anal ; 188: 113412, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32590301

ABSTRACT

Biopharmaceutical drug substances are generally produced using fermentation technology and are subsequently purified in the following downstream process. For the determination of critical quality attributes (CQAs), such as target protein titer and purity, monitoring tools are required before quality control analysis. We herein present a novel reversed phase liquid chromatography method (RPLC), which enables facile and robust protein quantification during upstream and downstream processing of intracellularly produced proteins in E. coli. The overall goal was to develop a fast, robust and mass spectrometry compatible method which can baseline resolve and quantify each protein of interest. Method development consisted of three steps, oriented on an Analytical Quality by Design (AQbD) workflow: (i) the stationary phase as primary parameter was chosen based on state-of-the art technology thus minimizing protein on-column adsorption and providing high efficiency, (ii) secondary parameters (i.e. gradient conditions and column temperature) were optimized applying chromatographic modeling, and (iii) the established Method Operable Design Region (MODR) was challenged and confirmed during robustness testing, performed in-silico and experimentally by a Design of experiment (DoE) based approach. Finally, we validated the RPLC method for pivotal validation parameters (i.e. linearity, limit of quantification, and repeatability) and compared it for protein quantification against a well-established analytical methodology. The outcome of this study shows (i) a protocol for RPLC development using an AQbD principle for new method generation and (ii) a highly versatile RPLC method, suited for quick and straightforward recombinant protein titer measurement being applicable for the detection of a broad range of proteins.


Subject(s)
Chromatography, Reverse-Phase , Escherichia coli , Chromatography, High Pressure Liquid , Mass Spectrometry , Quality Control , Research Design
8.
Int J Mol Sci ; 21(11)2020 May 29.
Article in English | MEDLINE | ID: mdl-32485932

ABSTRACT

The bacterium E. coli is one of the most important hosts for recombinant protein production. The benefits are high growth rates, inexpensive media, and high protein titers. However, complex proteins with high molecular weight and many disulfide bonds are expressed as inclusion bodies (IBs). In the last decade, the overall perception of these IBs being not functional proteins changed, as enzyme activity was found within IBs. Several applications for direct use of IBs are already reported in literature. While fluorescent proteins or protein tags are used for determination of IB activity to date, direct measurements of IB protein activity are scacre. The expression of recombinant hyaluronidase from Apis mellifera in E. coli BL21(DE3) was analyzed using a face centered design of experiment approach. Hyaluronidase is a hard to express protein and imposes a high metabolic burden to the host. Conditions giving a high specific IB titer were found at 25 °C at low specific substrate uptake rates and induction times of 2 to 4 h. The protein activity of hyaluronidase IBs was verified using (Fourier transform) FT-IR spectroscopy. Degradation of the substrate hyaluronan occurred at increased rates with higher IB concentrations. Active recombinant hyaluronidase IBs can be immediately used for direct degradation of hyaluronan without further down streaming steps. FT-IR spectroscopy was introduced as a method for tracking IB activity and showed differences in degradation behavior of hyaluronan dependent on the applied active IB concentration.


Subject(s)
Escherichia coli/metabolism , Hyaluronoglucosaminidase/biosynthesis , Inclusion Bodies/metabolism , Recombinant Proteins/biosynthesis , Animals , Bees , Biomass , Bioreactors , Culture Media/metabolism , Disulfides , Fermentation , Hyaluronic Acid/metabolism , Molecular Weight , Spectroscopy, Fourier Transform Infrared , Substrate Specificity , Temperature
9.
Pharmaceutics ; 12(4)2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32326286

ABSTRACT

Protein freeze-thawing is frequently used to stabilize and store recombinantly produced proteins after different unit operations in upstream and downstream processing. However, freeze-thawing is often accompanied by product damage and, hence, loss of product. Different effects are responsible, including cold denaturation, aggregation effects, which are caused by inhomogeneities in protein concentration, as well as pH and buffer ingredients, especially during the freeze cycle. In this study, we tested a commercially available small-scale protein freezing unit using immunoglobin G (IgG) as monoclonal antibody in a typical formulation buffer containing sodium phosphate, sodium chloride, and Tween 80. Different freezing rates were used respectively, and the product quality was tested in the frozen sample. Spatially resolved tests for protein concentration, pH, conductivity, and aggregation revealed high spatial differences in the frozen sample. Usage of slow freezing rates revealed high inhomogeneities in terms of buffer salt and protein distribution, while fast rates led to far lower spatial differences. These protein and buffer salt inhomogeneities can be reliably monitored using straight forward analytics, like conductivity and photometric total protein concentration measurements, reducing the need for HPLC analytics in screening experiments. Summarizing, fast freezing using steep rates shows promising results concerning homogeneity of the final frozen product and inhibits increased product aggregation.

10.
J Biotechnol ; 324S: 100022, 2020.
Article in English | MEDLINE | ID: mdl-34154734

ABSTRACT

Recombinant protein production in E. coli often leads to the formation of inclusion bodies (IBs). Although downstream processing of IBs has the reputation of being a great hurdle, advantages of IBs can be substantial. Highly pure recombinant protein with the possibility of correctly folded structures and an easy separation from cell matter are decisive factors that make IB processes so interesting. Product yield, purity and biological activity of the refolded protein are the responses to evaluate an IB process. The objective of this case study was to develop a refolding process in an integrated manner. The effects of the unit operations 1) homogenization, 2) IB wash and 3) IB solubilisation as well as their interdependencies were analyzed. We revealed interesting factor interactions between homogenization and IB wash, as well as homogenization and solubilisation, which would be overlooked if the single unit operations were investigated individually. Furthermore, we found that homogenization was a key unit operation for IB processing. By changing the conditions during homogenization only, the product yield, purity and biological activity of the refolded product was affected 2-fold, 1.2-fold and 2.5-fold, respectively.


Subject(s)
Escherichia coli , Inclusion Bodies , Recombinant Proteins
11.
Anal Bioanal Chem ; 412(9): 2081-2088, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31332469

ABSTRACT

Filamentous fungi are well-established production hosts that feature a strong interconnection between morphology, physiology, and productivity. For penicillin production in Penicillium chrysogenum, industrial processes frequently favor a pellet morphology comprising compact hyphal agglomerates. Inherently these tightly packed entanglements lead to inactive, degrading sections within the pellet's core because of limitations. Optimal process design requires detailed knowledge of the nature of the limitations and localization of productive zones in the biomass, which is generally obtainable through modeling and complex analytical methods such as oxygen microelectrode and histological investigations. Methods that combine physiological and morphological insight are crucial yet scarce for filamentous fungi. In this study, we used time-of-flight secondary ion mass spectrometry in combination with oxygen and glucose tracer substrates, requiring little effort for sample preparation and measurement. Our method is capable of analyzing oxygen and substrate uptake in various morphological structures by the use of 18O as a tracer. In parallel, we can assess productive biomass regions through identification of penicillin mass fragments to simultaneously study oxygen diffusion, substrate incorporation, and productive biomass sections.


Subject(s)
Penicillium chrysogenum/metabolism , Biomass , Fungi/growth & development , Fungi/metabolism , Glucose/metabolism , Oxygen/metabolism , Penicillins/metabolism , Penicillium chrysogenum/growth & development , Spectrometry, Mass, Secondary Ion
12.
Article in English | MEDLINE | ID: mdl-31824931

ABSTRACT

Escherichia coli still serves as a beloved workhorse for the production of many biopharmaceuticals as it fulfills essential criteria, such as having fast doubling times, exhibiting a low risk of contamination, and being easy to upscale. Most industrial processes in E. coli are carried out in fed-batch mode. However, recent trends show that the biotech industry is moving toward time-independent processing, trying to improve the space-time yield, and especially targeting constant quality attributes. In the 1950s, the term "chemostat" was introduced for the first time by Novick and Szilard, who followed up on the previous work performed by Monod. Chemostat processing resulted in a major hype 10 years after its official introduction. However, enthusiasm decreased as experiments suffered from genetic instabilities and physiology issues. Major improvements in strain engineering and the usage of tunable promotor systems facilitated chemostat processes. In addition, critical process parameters have been identified, and the effects they have on diverse quality attributes are understood in much more depth, thereby easing process control. By pooling the knowledge gained throughout the recent years, new applications, such as parallelization, cascade processing, and population controls, are applied nowadays. However, to control the highly heterogeneous cultivation broth to achieve stable productivity throughout long-term cultivations is still tricky. Within this review, we discuss the current state of E. coli fed-batch process understanding and its tech transfer potential within continuous processing. Furthermore, the achievements in the continuous upstream applications of E. coli and the continuous downstream processing of intracellular proteins will be discussed.

13.
Article in English | MEDLINE | ID: mdl-31737617

ABSTRACT

State of the art microbial recombinant protein production is regularly performed in fed-batch based cultivations. However, these cultivations suffer from highly time-dependent changes in productivity and product quality, leading to high variations in the downstream process. Continuous biomanufacturing offers the possibility of a time independent process, boosting the time-space-yield of the recombinantly produced protein and further reducing costs for production, also as downstream gets more predictive. In the current work, the continuous production of a pharmaceutically relevant protein in form of an inclusion body in E. coli BL21(DE3) was investigated in single vessel cultivations by varying dilution rates using glycerol as carbon source, inducer (lactose or IPTG) and respective inducer concentrations. Attempts to increase low specific productivities observed in single vessel continuous cultivations, led to the establishment of a continuously operated cascade of two stirred tank reactors to spatially separate biomass formation from recombinant protein production. Process performance was substantially improved compared to a single vessel chemostat culture, as specific productivity and space-time yield were boosted using an optimized cascaded process by about a factor of 100. This study shows the potential of a two-stage continuous process as promising alternative to benchmark fed-batch processes achieving constant inclusion body production at a time-independent level.

14.
J Biotechnol ; 296: 75-82, 2019 Apr 20.
Article in English | MEDLINE | ID: mdl-30904592

ABSTRACT

The Gram-negative bacterium E. coli is the host of choice for the production of a multitude of recombinant proteins in industry. Generally, cultivation is easy, media are cheap and a high product titer can be obtained. However, harsh induction procedures using IPTG as inducer are often referred to cause stress reactions, leading to a phenomenon known as metabolic burden and expression of inclusion bodies. In this contribution, we present different strategies for determination of critical timepoints for product stability in an E. coli IB bioprocess. As non-controlled feeding during induction regularly led to undesired product loss, we applied physiological feeding control. We found that the feeding strategy has indeed high impact on IB productivity. However, high applied qs,C increased IB product titer, but subsequently stressed the cells and finally led to product degradation. Calculating the cumulated glycerol uptake of the cells during induction phase (dSn), we found an empirical value, which serves as a strong indicator for process performance and can be used as process analytical tool. We tested different approaches starting from offline control. Glycerol accumulation could be used as trigger to establish a model-based approach to predict titer and viable cell concentration for a model protein. This straight forward control and model-based approach is high beneficial for upstream development and for increasing stability.


Subject(s)
Glycerol/chemistry , Inclusion Bodies/drug effects , Isopropyl Thiogalactoside/adverse effects , Recombinant Proteins/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Glycerol/metabolism , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Isopropyl Thiogalactoside/chemistry , Recombinant Proteins/biosynthesis
15.
Bioresour Technol ; 273: 277-287, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30448679

ABSTRACT

Recently, several mutants of Synechocystis sp. PCC 6714 were obtained showing superior PHB content and productivities. Here, the most promising mutant named MT_a24 is compared in detail with the wild-type in controlled photobioreactors. In order to provide an easily scalable and alternative approach to the normally done two-step process -comprising of growth phase and limitation phase- a one-step cultivation was optimized. The multivariate experimental design approach was used for the optimization of the one-step, self-limiting media. During one-step cultivation of MT_a24 with optimized media 30 ±â€¯4% (DCW) corresponding to 1.16 g L-1 PHB was obtained. Using pulse experiments it was demonstrated that phosphate is the key driver of glycogen synthesis in Synechocystis sp. PCC 6714 and it can be used to boost glycogen productivity. The maximum glycogen content acquired was 2.6 g L-1 (76.2% DCW) for mutant MT_a24 using phosphate feeding and carbon dioxide as carbon source.


Subject(s)
Carbohydrates/biosynthesis , Carbon Dioxide/metabolism , Photobioreactors , Synechocystis/metabolism , Carbohydrate Metabolism , Carbon/metabolism , Glycogen/biosynthesis
16.
Appl Microbiol Biotechnol ; 103(3): 1143-1153, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30569219

ABSTRACT

The bacterium Escherichia coli is a major host for recombinant protein production of non-glycosylated products. Depending on the expression strategy, the recombinant protein can be located intracellularly, which often leads to protein aggregates inside of the cytoplasm, forming so the called inclusion bodies (IBs). When compared to other protein expression strategies, inclusion body formation allows high product titers and also the possibility of expressing proteins being toxic for the host. In the past years, the comprehension of inclusion bodies being only inactive protein aggregates changed, and the new term of non-classical inclusion bodies emerged. These inclusion bodies are believed to contain a reasonable amount of active protein within their structure. However, subsequent downstream processing, such as homogenisation of cells, centrifugation or solubilisation of IBs, is prone to variable process performance and is often known to result in low extraction yields. It is hypothesised that variations in IB quality attributes are responsible for those effects and that such attributes can be controlled by upstream process conditions. In this review, we address the impact of process design (process parameters) in the upstream on defined inclusion body quality attributes. The following topics are therefore addressed: (i) an overview of the range of inclusion body applications (including emerging technologies); (ii) analytical methods to determine quality attributes; and (iii) screws in process engineering to achieve the desired quality attributes for different inclusion body-based applications. Process parameters in the upstream can be used to trigger different quality attributes including protein activity, but are not exploited to a satisfying content yet. Design by quality approaches in the upstream are already considered for a multitude of existing processes. Further intensifying this approach may pave the industrial application for new IB-based products and improves IB processing, as discussed within this review.


Subject(s)
Escherichia coli/metabolism , Inclusion Bodies/metabolism , Protein Engineering/methods , Recombinant Proteins/metabolism , Bacterial Physiological Phenomena , Cytoplasm/metabolism , Escherichia coli/genetics , Recombinant Proteins/genetics
17.
Microorganisms ; 6(4)2018 Nov 25.
Article in English | MEDLINE | ID: mdl-30477255

ABSTRACT

The Gram-negative bacterium E. coli is the host of choice for producing a multitude of recombinant proteins relevant in the pharmaceutical industry. Generally, cultivation is easy, media are cheap, and a high product titer can be obtained. However, harsh induction procedures combined with the usage of IPTG (isopropyl ß-d-1 thiogalactopyranoside) as an inducer are often believed to cause stress reactions, leading to intracellular protein aggregates, which are so known as so-called inclusion bodies (IBs). Downstream applications in bacterial processes cause the bottleneck in overall process performance, as bacteria lack many post-translational modifications, resulting in time and cost-intensive approaches. Especially purification of inclusion bodies is notoriously known for its long processing times and low yields. In this contribution, we present screening strategies for determination of inclusion body bead size in an E. coli-based bioprocess producing exclusively inclusion bodies. Size can be seen as a critical quality attribute (CQA), as changes in inclusion body behavior have a major effect on subsequent downstream processing. A model-based approach was used, aiming to trigger a distinct inclusion body size: Physiological feeding control, using qs,C as a critical process parameter, has a high impact on inclusion body size and could be modelled using a hyperbolic saturation mechanism calculated in form of a cumulated substrate uptake rate. Within this model, the sugar uptake rate of the cells, in the form of the cumulated sugar uptake-value, was simulated and considered being a key performance indicator for determination of the desired size. We want to highlight that the usage of the mentioned screening strategy in combination with a model-based approach will allow tuning of the process towards a certain inclusion body size using a qs based control only. Optimized inclusion body size at the time-point of harvest should stabilize downstream processing and, therefore, increase the overall time-space yield. Furthermore, production of distinct inclusion body size may be interesting for application as a biocatalyst and nanoparticulate material.

18.
Microb Cell Fact ; 17(1): 148, 2018 Sep 20.
Article in English | MEDLINE | ID: mdl-30236107

ABSTRACT

BACKGROUND: The bacterium E. coli is a major host for recombinant protein production of non-glycosylated products. Depending on the expression strategy, the recombinant protein can be located intracellularly. In many cases the formation of inclusion bodies (IBs), protein aggregates inside of the cytoplasm of the cell, is favored in order to achieve high productivities and to cope with toxic products. However, subsequent downstream processing, including homogenization of the cells, centrifugation or solubilization of the IBs, is prone to variable process performance or can be characterized by low extraction yields as published elsewhere. It is hypothesized that variations in IB quality attributes (QA) are responsible for those effects and that such attributes can be controlled by upstream process conditions. This contribution is aimed at analyzing how standard process parameters, such as pH and temperature (T) as well as different controlled levels of physiological parameters, such as specific substrate uptake rates, can vary IB quality attributes. RESULTS: Classical process parameters like pH and T influence the expression of analyzed IB. The effect on the three QAs titer, size and purity could be successfully revealed. The developed data driven model showed that low temperatures and low pH are favorable for the expression of the two tested industrially relevant proteins. Based on this knowledge, physiological control using specific substrate feeding rate (of glucose) qs,Glu is altered and the impact is tested for one protein. CONCLUSIONS: Time dependent monitoring of IB QA-titer, purity, IB bead size-showed a dependence on classical process parameters pH and temperature. These findings are confirmed using a second industrially relevant strain. Optimized process conditions for pH and temperature were used to determine dependence on the physiological parameters, the specific substrate uptake rate (qs,Glu). Higher qs,Glu were shown to have a strong influence on the analyzed IB QAs and drastically increase the titer and purity in early time stages. We therefore present a novel approach to modulate-time dependently-quality attributes in upstream processing to enable robust downstream processing.


Subject(s)
Escherichia coli , Inclusion Bodies/metabolism , Cytoplasm/metabolism , Hydrogen-Ion Concentration , Metabolic Engineering/methods , Protein Aggregates , Recombinant Proteins/biosynthesis , Temperature
19.
Appl Microbiol Biotechnol ; 102(2): 667-676, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29159587

ABSTRACT

Against the outdated belief that inclusion bodies (IBs) in Escherichia coli are only inactive aggregates of misfolded protein, and thus should be avoided during recombinant protein production, numerous biopharmaceutically important proteins are currently produced as IBs. To obtain correctly folded, soluble product, IBs have to be processed, namely, harvested, solubilized, and refolded. Several years ago, it was discovered that, depending on cultivation conditions and protein properties, IBs contain partially correctly folded protein structures, which makes IB processing more efficient. Here, we present a method of tailored induction of recombinant protein production in E. coli by a mixed feed system using glucose and lactose and its impact on IB formation. Our method allows tuning of IB amount, IB size, size distribution, and purity, which does not only facilitate IB processing, but is also crucial for potential direct applications of IBs as nanomaterials and biomaterials in regenerative medicine.


Subject(s)
Escherichia coli/metabolism , Inclusion Bodies/metabolism , Recombinant Proteins/biosynthesis , Culture Media , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Glucose/metabolism , Inclusion Bodies/genetics , Lactose/metabolism , Protein Folding , Recombinant Proteins/genetics , Solubility
20.
Bioengineering (Basel) ; 5(1)2017 Dec 21.
Article in English | MEDLINE | ID: mdl-29267215

ABSTRACT

The Gram-negative bacterium E. coli is the host of choice for a multitude of used recombinant proteins. Generally, cultivation is easy, media are cheap, and a high product titer can be obtained. However, harsh induction procedures using isopropyl ß-d-1 thiogalactopyranoside as inducer are often referred to cause stress reactions, leading to a phenomenon known as "metabolic" or "product burden". These high expressions of recombinant proteins mainly result in decreased growth rates and cell lysis at elevated induction times. Therefore, approaches tend to use "soft" or "tunable" induction with lactose and reduce the stress level of the production host. The usage of glucose as energy source in combination with lactose as induction reagent causes catabolite repression effects on lactose uptake kinetics and as a consequence reduced product titer. Glycerol-as an alternative carbon source-is already known to have positive impact on product formation when coupled with glucose and lactose in auto-induction systems, and has been referred to show no signs of repression when cultivated with lactose concomitantly. In recent research activities, the impact of different products on the lactose uptake using glucose as carbon source was highlighted, and a mechanistic model for glucose-lactose induction systems showed correlations between specific substrate uptake rate for glucose or glycerol (qs,C) and the maximum specific lactose uptake rate (qs,lac,max). In this study, we investigated the mechanistic of glycerol uptake when using the inducer lactose. We were able to show that a product-producing strain has significantly higher inducer uptake rates when being compared to a non-producer strain. Additionally, it was shown that glycerol has beneficial effects on viability of cells and on productivity of the recombinant protein compared to glucose.

SELECTION OF CITATIONS
SEARCH DETAIL
...