Practice 2: Recombinant proteins: from small- to large-scale expression technologies Location: Loschmidt Laboratories, Kamenice 5/A13, 2nd floor, room 332 Lecturers: Ing. RNDr. Martin Marek, Ph.D. (martin.marek@recetox.muni.cz) Ing. Andrea Schenkmayerova, Ph.D. (andrea.schenkmayerova@fnusa.cz) I. WORKFLOW Selection of an expression host - purpose of protein production - combinatorial screening in small-scale formats - bench-scale and large-scale (fermentor) overproductions - downstream processing and purification strategies - quality/quantity check and final formulation II. MOTIVATION Proteins are produced in heterologous systems because of the impossibility to obtain satisfactory yields from natural sources. The production of soluble and functional recombinant proteins is among the main goals in the biotechnological and pharmaceutical industries. The selection of an optimal expression organism (host) and the most appropriate growth conditions to minimize the formation of insoluble proteins have to be done according to the protein characteristics and downstream requirements. Escherichia coli is the most popular recombinant protein expression system despite the great development achieved so far by eukaryotic expression systems. However, it is important to mention that E. coli expression system possesses, in many cases, severe limitations for a successful recombinant protein production. Therefore, eukaryotic systems, including mammalian cells, insect cells, yeasts, filamentous fungus, and microalgae, are an important alternative for the production of those difficult-to-express proteins. During this course we will combine theoretical designs with practical demonstrations concerning all aspects of recombinant protein (multi)-expressions, ranging from small- to large-scale (fermentor) overproduction technologies. In the second part, we will focus on the downstream processing and purification strategies. The students will get knowledge on: - How to select a heterologous system (expression host) - Pilot small-scale (transient) expression screening to determine optimal expression conditions, and buffer screening strategies - Scaling-up of production process, bench-scale and large-scale (fermentor) productions - Harvesting process and timing - Downstream processing and purification strategies to yield highly pure recombinant proteins in sufficient amounts - Control (quality/quantity) of the purified protein, final formulation and long-term storage III. THEORETICAL BACKGROUND Selection of expression organism (host): each heterologous expression system has benefits and drawbacks with respect to their capacity for recombinant protein production (Figure 1). The gram-negative bacterium Escherichia coli is frequently the first expression host chosen for the production 1 of a recombinant protein, owing to the rapid, affordable and technically straightforward culturing associated with its use. The E. coli system offers a mean for rapid, high yield, and economical production of recombinant proteins (Figure 2). However, high-level production of functional eukaryotic proteins in E. coli may not be a routine matter, sometimes it is quite challenging. Techniques to optimize heterologous protein overproduction in E. coli have been explored for host strain selection, plasmid copy numbers, promoter selection, mRNA stability, and codon usage, significantly enhancing the yields of the foreign eukaryotic proteins. Yeasts is a single-celled eukaryotic organism capable of producing very large quantities of recombinant protein. Pichia pastoris is the most used strain of first choice for yeast expression. The baculovirus/insect cell expression vector system (BEVS) is a popular choice for the production of recombinant proteins, particularly those requiring complex post-translational modifications or integral membrane proteins. Mammalian cell-based expression (e.g. HEK293 cell line) is the dominant system for the production of therapeutic recombinant proteins. Their capacity to handle complex post-translational modifications, folding and assembly of recombinant proteins and protein complexes is superior to other systems. The Leishmania tarentolae extract (LTE) in vitro translation cell-free expression system is a rapid, convenient, flexible and cost effective tool to produce recombinant proteins for biochemical, biophysical and structural analysis. LOW HIGH SPEED MAMMALIAN ESE.VS/IN5ECT CELL YEAST BACTERIA COST BACTERIA I!© Y-EA5T BEUS/IN5ECT CELL MAMMALIAN TYPICAL YIELD MAMMALIAN BEVS/INSECT CELL BACTERIA YEAST POST-TRANSLATION MODIFICATION BACTERIA TEAST BEYS/INSECT CELL MAMMALIAN FDA APPROVAL Figure 1. Comparison of the most industrially used expression systems. eivyiwiECTCEn MAMMALIAN At the beginning of a protein expression project, small-scale expression screenings in minimal volumes (2-6 mL) are recommended prior to running large-scale overproductions. The pilot small-scale expression tests are designed to test different expression parameters (host strain selection, timing of induction and harvesting, expression temperature, pH, ionic strength etc.) to find an optimal condition and buffer screening. Large-scale expression in bacteria, yeast, insect and mammalian cell systems in culture volumes from 400 mL to 10 L are available in shake flask and from 2 L to 20 L in a stirred-tank bioreactor or up to 25 L in a Wave bioreactor. Large-scale production in bioreactor (fermentor) is very complex procedure where several factors play a crucial role on the performance of the culture. Composition of cultivation broth, sterilization efficiency, proper agitation and aeration, temperature or feeding strategy can all together influence final yield of biomass and recombinant protein from the fermentation. Since culture in fermentor is half opened system, it is very susceptible to contamination. It is very important for the operator of the fermentor to maintain sterility throughout the fermentation. In this practice, the students will learn how to assemble the fermentor vessel and prepare it for sterilization, how to calibrate pH and DO (dissolved oxygen) sensors and set up the fermentation together with creation of recipes for fed-batch cultivations. 2 Recombinant protein expression in E. coli: basic principles and advantages versus limitations ADVANTAGES Inexpensive setup and running costs High recombinant protein production levels Short timeline from cloning to protein recovery (1 week) Limited technical knowledge required for culturing Scalability from small (2 mL) to very large industrial culture (>10,000 L) volumes DISADVANTAGES Inability to perform post-translation modifications (PTMs) Limited formation of disulphide bond Figure 2. Schematic representation of the plasmid used for the protein expression in E. coll. (A) Map of the typical expression plasmid. The gene of interest (DAC2) is inserted between A/del and BamH\ restriction sites. Selection in E. coli is performed by the beta-lactamase ampicillin resistance gene (ampR). Origin of replication sequence (ColEl) is available for maintenance in F. coli cells, and the lacl gene is present for expression of the Lac repressor protein. (B) Details of the expression cassette. The gene of interest (DAC2) is controlled by the 17 promoter and 17 terminator. The DAC2 gene is in cloned in frame with a sequence coding for a C-terminal thrombin cleavage site followed by a poly-histidine tag. (C) Overview of recombinant protein expression workflow in F. coli. Recombinant protein purification: the aim of a purification procedure is to obtain a highly pure and stable protein at an appropriate concentration in a buffer compatible with the intended downstream application (Figure 3). Chromatographic techniques are the most powerful and commonly used means of purifying recombinant proteins. Each technique separates proteins based on different properties, so it is often advantageous to combine several types to maximise separation of the 3 recombinant protein from host cell proteins. The use of fusion tags (polyhistidines, Strep-tag, GST, MBP, SUMO, thioredoxin etc.) not only facilitates affinity chromatography steps, but also can dramatically improve protein expression, stability, resistance to proteolytic degradation and solubility. Variety of proteases (thrombin, protease 3C, TEV etc.) for fusion tag removal during downstream processing is available. Recombinant protein expression 1 Clone V J Transform Culture Lyse cell pellet (Sonication, high-pressure homogenizer, Microfluidizers) Filter Chromatography steps Affinity chromatography Ion-exchange chromatography Size-exclusion chromatography Hydrophobic interaction chromatography Reversed phase chromatography Filtration Clarify cell lysate by high-speed centrifugation Figure 3. Workflow of recombinant protein purification includes (i) the lysis of a cell pellet by sonication or high-pressure homogenizer, (i) the clarification of cell lysate by high-speed centrifugation, (iii) filtration, and (iv) one or more chromatographic purification steps. Recombinant protein characterization: an aggregation problem. A key challenge in recombinant protein production is to maintain and store the target protein in a soluble and stable form. Protein aggregation can compromise protein function and thus it is necessary to overcome this challenge to Protein aggregation Monomer ooo OOq0 It o obs.Mw-100 n ■ c ■ I where 6>0bs is the observed ellipticity in degrees, Mw is the protein molecular weight, n is number of residues, / is the cell path length, c is the protein concentration (in mg/ml) and the factor 100 originates from the conversion of the molecular weight to mg/mol. 3. Dynamic light scattering (DLS): The dynamic light scattering (DLS) experiments are conducted typically with protein solutions (l-2mg/ml) in a corresponding buffer containing using instrument DynaPro NanoStar (Wyatt). Protein solutions are centrifuged (13,000 rpm/10 min) prior to DLS measurement in order to remove impurities. Before measurement temperature is equilibrated to 20°C. 4. Differential scanning fluorimetry (nanoDSF) measurements: Thermal stability of recombinant proteins is analysed by a label-free differential scanning fluorimetry (DSF) approach using a Prometheus NT.48 instrument (NanoTemper Technologies). Briefly, the shift of intrinsic tryptophan fluorescence of proteins upon gradual temperature-triggered 9 unfolding (temperature gradient 20-95°C) is monitored by detecting the emission fluorescence at 330 and 350 nm. The measurements is carried out in nanoDSF-grade high sensitivity glass capillaries (NanoTemper Technologies) at a heating rate of l°C/min. Protein melting points (Tm) are inferred from the first derivative of the ratio of tryptophan emission intensities at 330 and 350 nm. VI. LITERATURE RosanoG. L. and Ceccarelli E. A. (2014) Recombinant protein expression in Escherichia coli: advances and challenges. Frontiers in Microbiology. 5: 172. Young C. L, Britton Z. T., Robinson A. S. (2012) Recombinant protein expression and purification: a comprehensive review of affinity tags and microbial applications. Biotechnology Journal. 7: 620-34. Sivashanmugam A., Murray V., Cui C, Zhang Y., Wang J., Li Q. (2009) Practical protocols for production of very high yields of recombinant proteins using Escherichia coli. Protein Science: 18(5): 936-48. Diebold M-L, Fribourg S, Koch M, Metzger T, Romier C. (2011) Deciphering correct strategies for multiprotein complex assembly by co-expression: Application to complexes as large as the histone octamer. Journal of Structural Biology 175(2), 178-188. Formenti, L. R., N0rregaard, A., Bolic, A., Hernandez, D. Q., Hagemann, T., Heins, A.-L., Larsson, H., Mears, L., Mauricio-lglesias, M., Krtihne, U., Gernaey, K. V. (2014) Challenges in industrial fermentation technology research. Biotechnol. J., 9: 727-738. Kuprijanov, A., Schaepe, S., Aehle, M., Simutis, R., Liibbert, A. (2012) Improving cultivation processes for recombinant protein production. Bioprocess Biosyst. Eng. 35: 333-340. V. HOMEWORK Your goal is to recombinantly produce of 2 g of highly pure haloalkane dehalogenase DhaA enzyme from Desulfobacterium autotrophicum. See amino acid and corresponding nucleotide sequences below: >ACN15444.1 DhaA [Desulfobacterium autotrophicum HRM2] MVTRDPAEQSRNIKSPGIRRKINGTMVGTKDFYEIYPFVPHFMTLDRHKLHYLDLGKGSPWMVHGNPTWSFYFRRLARDLSV NHRVIVPDHMGCGLSDKPSTRDYDYTLASRVRDLDRLIQSLDLGKKITLVVHDWGGMIGCAWALRHLDRIDRIIITNTSGFHL PGAKRFPLRLWLIKYLPWFAIPGIQGLNLFAPAALYMAPKQSLSTTVRQGLTAPYNSWKNRIATLKFVQDIPLSPRDKSYELV NWVDTHLEGLKTVPMMILWGRHDFVFDLSFLDEWNKRFPHAQTHIFEDAGHYLFEDKPDETSNLIKKFIEEY >CP001087.1:2679075-2680040 Desulfobacterium autotrophicum HRM2, complete genome ATGGTAACCAGGGATCCAGCGGAGCAAAGCAGAAACATCAAAAGTCCGGGCATCAGAAGAAAGATCAACGGCACCATGGTCGG CACCAAGGATTTTTATGAAATATATCCCTTTGTTCCCCATTTCATGACCCTGGACCGGCACAAACTCCACTACCTTGACCTGG GTAAGGGAAGTCCAGTTGTCATGGTCCACGGTAATCCCACCTGGTCGTTTTATTTTCGCAGGCTTGCCCGGGATCTTTCGGTG AACCACCGGGTCATTGTTCCCGACCACATGGGGTGCGGCCTGTCTGACAAGCCGTCCACCAGGGATTACGACTATACCCTTGC ATCAAGGGTCCGGGACCTGGACCGTCTGATCCAGAGCCTTGACCTTGGAAAAAAGATCACCCTGGTCGTCCACGACTGGGGCG GTATGATCGGCTGCGCCTGGGCCCTTCGTCACCTGGACAGGATAGACAGGATCATCATCACCAACACCTCGGGGTTTCATCTT CCCGGGGCAAAACGATTTCCCCTGCGGCTTTGGCTGATCAAATACCTTCCCTGGTTTGCCATTCCAGGGATTCAGGGCCTGAA TCTCTTTGCCAGGGCAGCCCTTTACATGGCTCCGAAACAATCACTTTCAACAACGGTCAGGCAGGGGCTCACGGCACCCTACA ACTCGTGGAAAAACAGGATCGCCACCCTCAAATTTGTCCAGGACATTCCCCTTTCACCCAGGGACAAAAGCTACGAACTTGTC AACTGGGTGGACACCCACCTTGAAGGTCTTAAAACCGTTCCCATGATGATCCTATGGGGCAGACACGATTTTGTGTTTGATCT GTCGTTCCTTGACGAGTGGAACAAACGGTTTCCCCATGCCCAAACACATATTTTCGAGGATGCAGGCCATTATCTGTTTGAGG ACAAAC C C GAT GAAACAT CAAAT C T TAT CAAAAAAT T CATAGAG GAG TACTAA 10 Select a suitable expression host (heterologous system) for the DhaA enzyme overproduction and explain why the selected host is the best choice: Propose and design a strategy for the DNA template synthesis, including primer design: Propose a cloning strategy - ligation-dependent versus ligation-independent cloning, selection of expression vector, affinity/solubility tags etc.? How will you check the error-free clones? 4. Briefly describe production process - how will you introduce a foreign gene into the host, from pre-culture to large-scale overproduction, inducible versus stable expression, cytotoxicity issue, timing, harvesting strategy etc. 5. How will you determine the quality and yield of the purified enzyme? 6. How will you determine oligomeric state of the DhaA enzyme? 12