BioMedNet HomeLibraryDatabasesCollaborationsJob ExchangeShopping MallYour Room


FULL TEXT (TEXT + FULL FIGURES)

Current Opinion in Biotechnology
Vol. 7, No. 5, October 1996
The expression of recombinant proteins in yeasts
[Review article]
Peter E Sudbery
Current Opinion in Biotechnology 1996, 7:517-524.
 
Viewing options [Help]

text only | + thumbnails | + full figures
 
 
Associated links
 
 
-> Publications by
Peter E Sudbery
 
 
-> Jump to this record in Evaluated MEDLINE
 
 
-> Related records from Evaluated MEDLINE
 
 
-> Related fulltext articles on BioMedNet
 
 
-> Fulltext articles on BioMedNet that cite this article
 


Outline


Abstract
The methylotrophic yeasts Hansenula polymorpha and Pichia pastoris are rapidly becoming the systems of choice for the expression of recombinant proteins in yeast. However, the powerful genetic techniques available in Saccharomyces cerevisiae and the fission yeast Schizosaccharomyces pombe are still exploited to establish models to study medically important cell processes and screen for pharmacologically active compounds.

Abbreviations
ABC—ATP-binding cassette;
AOX—alcohol oxidase;
FGF—fibroblast growth factor;
HMG2/1—3-hydroxy-3-methylglutaryl-CoA reductase isoenzymes 1 and 2;
IFN-tau—interferon-tau;
MAPK—mitogen-activated protein kinase;
MF—mating factor;
MOX—methanol oxidase;
MRP—multidrug resistance associated protein;
OR—oxidoreductase;
PTS—peroxisomal targeting sequence;
rHbA—recombinant haemoglobin;
SSTR2—somatostatin receptor subtype 2.


Introduction

The advantages of yeast as hosts for the expression of recombinant proteins from higher eukaryotes have long been appreciated (for a general review see [1]). They combine the ease, simplicity and cheapness of bacterial expression systems with the authenticity of the far more expensive and less convenient animal tissue culture systems. Like bacteria, yeast are simple to cultivate on inexpensive growth media, and there is a formidable array of techniques for the manipulation of foreign genes. However, as eukaryotes, they provide an environment for post-translational processing and secretion, resulting in a product that is often identical or more similar to the native protein. Moreover, the budding yeast Saccharomyces cerevisiae has a long history of use in the production of bread and alcoholic beverages. As a result, there is confidence that the organism is safe, and experience of industrial-scale fermentations facilitates production scale-up.

The first yeast to be employed for the production of recombinant proteins was S. cerevisiae. Foreign genes are either maintained on 2 µm-based plasmids, which maintain high copy number but require selection upon growth in large fermenters or in a low copy integrated form. Strong, unregulated promoters such as that from the PGK1 gene, or the regulatable GAL1 promoter, may be employed. A particular advantage of yeast is that the foreign protein may be directed into the secretory pathway, usually by fusion of the mature form of the recombinant protein to the prepro sequence of the mating factor (MF). The pro sequence is removed by the proteolytic action of the Kex2 enzyme, a processing step that is widely conserved in eukaryotes. During secretion, protein folding, disulphide bond formation and glycosylation takes place. N-linked glycosylation occurs at the correct sites but often differs from the native pattern in that the carbohydrate chains are often of excessive length and are of the high-mannose type. The resulting protein may therefore be immunogenic and this limits the use of glycosylated proteins as human therapeutics. This basic methodology was established by the mid 1980s and led to the successful production of the hepatitis B vaccine based on recombinant HBsAg (hepatitis B surface antigen). Recombinant human serum albumin (rHA) produced in S. cerevisiae is also in clinical trials as a plasma expander [2]. Native serum albumin contains 17 disulphide bonds but is not glycosylated. The protein is produced by yeast in completely authentic from. To be economic, it must be produced in tonne quantities at US$2–3 g-1. This will necessitate the construction of a fermentation facility of the order of 100 m3. Such an undertaking is testament to the ability of yeast to provide the dual advantages of authenticity at low cost.

Despite these successes, S. cerevisiae is often limited as an expression system by low yields. Methylotrophic yeasts such as Pichia pastoris and Hansenula polymorpha retain all the advantages of S. cerevisiae but provide a reliable means of achieving greatly elevated yields [3] [4•]. Moreover, P. pastoris may, at least partially, solve the glycosylation problem. Both H. polymorpha and P. pastoris can grow on simple, defined media utilizing methanol as the sole carbon source. The first enzyme in the methanol-utilization pathway is an alcohol (methanol) oxidase known as AOX in P. pastoris and MOX in H. polymorpha. During growth on methanol, this enzyme constitutes some 30% of the total cell protein, sequestered in peroxisomes that can occupy 80% of total cell volume. The expression of AOX/MOX is highly regulated. In both H. polymorpha and P. pastoris, the enzyme is repressed by glucose or ethanol so that it is undetectable. In both yeasts, full induction requires the presence of methanol. In H. polymorpha, but not P. pastoris, MOX is derepressed to 10–20% of the maximum during growth on a nonrepressing carbon source such as glycerol. The powerful, regulatable promoter that programs the expression of AOX/MOX has been fully exploited in both yeasts for the production of recombinant proteins. The expression cassette is normally integrated by transplacement or duplicative integration and is stable during nonselective growth. A variety of approaches are available to increase copy number, which, in some cases, can increase yield. Fermentation in a bioreactor is essential for high expression levels but useful quantities of protein can be obtained from shake flasks. Fermentation protocols are simple: very high dry-cell weights can readily be obtained and biomass production can be separated from the production phase.

The expression of a heterologous gene in yeast may be undertaken because the recombinant protein has intrinsic commercial value, or the engineered strain may serve as the basis to develop other products. This may come about through the study of a cellular process of medical importance, an understanding of which may ultimately lead to the development of novel therapeutics. For example, the recombinant protein may be used for structural studies leading ultimately to the rational design of a small chemical inhibitor or activator; alternatively, the engineered strain may serve as the basis for a drug screening program. Alternatively, a mammalian receptor may be expressed, so that a screen can be established to search for drugs that act as agonists or antagonists. In the latter case, the similarity of fundamental cellular processes between yeast and higher organisms, coupled with the highly developed genetics of S. cerevisiae, make this yeast an extremely powerful experimental system.

This review will be concerned with examples, published since January 1995, of recombinant proteins of biotechnological relevance that have been expressed in yeast. For convenience, proteins of intrinsic value will be separated from those where the interest lies in the construction of model systems. The technology is now essentially mature and there have been no revolutionary developments. However, useful contributions to the technical side have appeared and these will also be considered.



Recombinant proteins of intrinsic value


Production in P. pastoris of small proteins uniformally labelled with 13C for two-dimensional NMR spectroscopy

Production of such proteins in S. cerevisiae is limited by the cost of 13C-labelled glucose. The use of 13C-labelled methanol reduces the cost tenfold, and could make the technique routine where previously it was applicable only in the most exceptional circumstances. The utility of this idea was demonstrated by the expression of tick anticoagulant peptide (TAP) from the soft tick Ornithodoros moubata in P. pastoris [5]. The 60 amino acid TAP polypeptide is a potent and specific inhibitor of blood coagulation factor Xa and so could be an important therapeutic agent. A synthetic, codon-optimized gene was expressed in P. pastoris fused with the S. cerevisiae MF prepro sequence to direct secretion. A yield of 1.7 gl-1 was obtained from a single copy insertion. In molar terms, this represents the highest yield reported from P. pastoris and was a sevenfold increase in productivity compared to S. cerevisiae. This system was used to produce single and dual 13C-thinspace and 15N-labelled protein enriched for more than 98% as determined by one-dimensional NMR.



Hirudin

Hirudin is a potent thrombin inhibitor produced by the leech Hirudo medicinalis. It has been successfully expressed in both S. cerevisiae and H. polymorpha [6] [7] [8•] [9•] [10] [11] [12] [13]. The highest yield was reported in H. polymorpha using a variety of leader sequences. Yields in the gml-1 range were reported [13]. A second group made the interesting observation that yields were greatly elevated by the addition of soybean oil to the medium, which aids yeast growth by providing essential fatty acids [9•]. In S. cerevisiae, a vector based on the use of the copper-inducible metallothionein (CUP1) gene has been used [8•]. This allows selection in complex medium by the addition of cupric sulphate. In copper-sensitive hosts, the apparent plasmid stability was 100%. The selection resulted in a twofold increase in plasmid copy number. The highest yield was obtained using the CUP1 promoter to program the expression of hirudin. In such a situation, the transcription of hirudin is directly linked to the transcription of the selectable marker, so an increase in selective pressure leads directly to an increase in product yield.



Antibodies and antibody receptors

Recombinant antibodies are likely to be one of the major products of the biotechnology industry. The use of murine monoclonal antibodies for diagnostic purposes is well known, but their use as therapeutic agents is limited by the likelihood of an immune response in a human subject. Monoclonal human antibodies can be produced by the powerful combinatorial library approach. Improvements in specificity and avidity can be made by clonal selection using phage display systems. Rational engineering can be used to produce novel proteins that, for example, may link a toxin or radioisotope to an antibody raised against a tumour antigen. Such antibodies may be produced in bacteria using signal sequences to direct secretion to the periplasmic space; however, yields are often disappointingly low. P. pastoris has been used to efficiently express rabbit scFv antibody fragments that recognize human leukaemia inhibitory factor isolated from a combinatorial library [14••]. Single chain (sc) Fv fragments consist of VL and VH domains joined by a short linker. The protein was tagged with five consecutive histidine residues to aid purification by Ni+-chelate affinity chromatography and secretion was directed using the S. cerevisiae MF prepro sequence. Shake flask cultures produced 100 mgl-1 of active protein, a 100-fold increase compared to E. coli. Further increases in yield may be expected when a fermenter is used.

As well as antibodies, antibody receptors may be expressed in P. pastoris (S Cain, B Helm, personal communication). Human IgE mediates the allergic response. It binds to two receptors, both of which are involved in type 1 hypersensitivity reactions. The high affinity receptor (FcRI) is found on mast cells and basophils and mediates the immune responses following cross-linking to IgE at the cell surface. It is a tetrameric protein of the form alphabetagammagamma. The extracellular portion of the alpha subunit binds IgE. It contains two immunoglobulin-like domains, the second of which binds IgE, and the first confers high affinity to the binding reaction. The extracellular domain of the chain has been expressed in P. pastoris both in its native form and as the individual immunoglobulin-like domains. After growth in a fermenter, 200–250 mgl-1 of product was obtained from the strain expressing the first domain, and 60 mgl-1 from each of the strains expressing either the second domain or the total extracellular domain. In all cases, the fragments were secreted into the culture medium. Previous attempts to express these fragments in E. coli and Baculovirus were disappointing, demonstrating the efficacy of this yeast system. IgE binding to the recombinant FcRI was examined using a BIAcore Ambis scanner and found to be indistinguishable from the native reaction. The structure of the receptor may now be studied by two-dimensional NMR after isotopic labelling of the proteins using 13C-labelled methanol as discussed above. The IgE constant region has also been expressed so that the structure of the IgE–receptor complex may be studied using X-ray crystallography.



Bovine enterokinase

Protein fusions are commonly employed in many different expression systems. Yield and biological activity of the recombinant protein may be increased by fusion to a rapidly folding protein such as thioredoxin to aid folding and enhance solubility, or, alternatively, to a peptide tag recognized by a commercially available monoclonal antibody or to histidine residues for binding to an Ni+-chelate affinity chromatography column. Whatever the reason for the fusion, an authentic protein requires cleavage of the chimeric protein. This is usually achieved by including a short peptide linker between the two moieties that forms a target for a protease with high specificity. Because of the increasing use of such fusions, there is an important market for the high specificity proteases. One such protease is bovine enterokinase, a serine protease that is the physiological activator of trypsinogen. The target of enterokinase is a Lys–Ile dipeptide that is preceded by a negatively charged Asp4 sequence. Enterokinase is a heterodimeric protein, consisting of a 115 kDa structural subunit that anchors a 35 kDa catalytic subunit to the intestinal brush border. The recombinant catalytic subunit produced in COS cells or E. coli retains the specificity of the holoenzyme but exhibits a 25-fold increase in activity. Shake flask cultures of engineered P. pastoris produced 6.5 mgl-1 of enzyme [15]. This enzyme was used to process thioredoxin–CAT and calmodulin–GFP fusion proteins produced in E. coli (CAT, chloramphenicol transacetylase; GFP, green fluorescent protein) and was found to be 100-fold more reactive than the native holoenzyme. Although the yield is low in comparison with other proteins, it is enough to process 1.7 g of CAT or 286.4 g of GFP fusions!



Recombinant antigens for use in vaccines

The Bm86 antigen from the cattle tick Boophilus microplus has been expressed in P. pastoris [16]. The recombinant protein was glycosylated and formed particles 17–45 nm in diameter which proved to be highly immunogenic, so that ticks engorging on vaccinated cattle were significantly damaged. An amino-terminally truncated form of outer-surface protein A from the Lyme disease spirochete Borrelia burgdorferi has been expressed in S. cerevisiae, resulting in a yield of 2.5 gl-1 [16]. Attempts to express this protein in E. coli had proven unsuccessful. Finally, Pfs25, a cysteine-rich 25 kDa protein present on the surface of Plasmodium falciparum zygotes has been expressed in S. cerevisiae [17]. When the recombinant protein was adsorbed to alum, it induced antibodies in both rodents and primates. These antibodies blocked the transmission of malaria parasites when mixed with infectious blood and fed to mosquitoes through a membrane feeding apparatus. Furthermore, unlike monoclonal antibodies to Pfs25, which block transmission only after ookinete development, antisera to Pfs25-B adsorbed to alum appeared to block the in vivo development of zygotes to ookinetes as well.



Interferon-tau

Interferon-tau (IFN-tau) signals the presence of an early conceptus to the maternal uterus and ensures the continued production of progesterone by the ovarian corpus luteum. 280 mgl-1 of IFN-tau was secreted in a sufficiently pure form such that, for many purposes, no further purification is necessary.



Stabilization and peroxisomal targeting of small peptides

Small peptides can be difficult to express in yeast. However, expression of human insulin-like growth factor II (IGFII; 67 amino acids) and Xenopus laevis magainin (23 amino acids) was found to be facilitated in H. polymorpha by fusion to the carboxyl terminus of amine oxidase [18••]. High-level synthesis of the fusion proteins, exceeding 20% of total cell protein, was obtained in methanol-limited chemostat cultures. After synthesis, the peptides could be readily separated from amine oxidase through factor Xa cleavage of a target sequence engineered into the linker. Although amine oxidase is normally targeted to the peroxisome via the PTS2 (PTS, peroxisomal targeting sequence) import pathway, the fusion proteins remained cytosolic. However, the addition of a carboxy-terminal SKL (PTS1) sequence to the AMO–IGFII construct did direct the recombinant protein to the peroxisome. This is an important finding, because it demonstrates a novel route for the production of recombinant proteins in methylotrophic yeasts. Peroxisomal targeting may reduce any toxic effects of the recombinant protein and, because peroxisomes can readily be separated from cell debris, it may facilitate the recovery and purification of an internally expressed protein.



Model systems


Transmembrane receptors

As well as providing products of direct value, the expression of proteins in yeast can be used to establish experimental systems for the development of other pharmaceutical products. For example, the expression of mammalian receptor proteins in yeast can form the basis of screens for small chemical drugs that may act as agonists or antagonists of receptor function. An important and elegant example of this is provided by the expression of rat somatostatin receptor subtype 2 (SSTR2) in S. cerevisiae [19••] (Fig. 1). This is a G-protein-coupled receptor with seven transmembrane domains. G-proteins are heterotrimeric proteins consisting of alpha, beta and gamma subunits. The alpha subunit binds GTP and interacts with both the receptor and intracellular effectors and so defines the activity of the G-protein complex. Yeast mating pheromones also bind to G-protein-coupled receptors with seven transmembrane domains. Upon pheromone binding, the trimeric complex dissociates, relieving negative regulation by the subunit on the dimer. This dimer then stimulates a mitogen-activated protein kinase (MAPK) cascade, which has two consequences: firstly, the activation of FAR1 and consequent cell cycle arrest in G1; and secondly, the transcriptional activation of FUS1. In order to create a yeast cell that can respond to somatostatin activation, the following changes were engineered in the his3 host: firstly, SSTR2 was expressed from a GAL1 promoter on a multicopy plasmid; secondly, the FAR1 gene was deleted, preventing cell-cycle arrest upon activation of the mating pheromone pathway; thirdly, the FUS1 gene was replaced with a fusion of the FUS1 pheromone responsive elements to the HIS3 coding sequences, so making His3 expression dependent upon activation of the mating pheromone pathway; fourthly, the GPA1 gene encoding the yeast Galpha subunit was deleted, to be replaced by a gene fusion consisting of the amino-terminal Gbetagamma interacting domain from GPA1 and the carboxy-terminal receptor-interacting domain of the rat Galphai2, thus allowing recombinant SSTR2 to stimulate the yeast pheromone response pathway; and fifthly, the yeast SST2 gene was deleted, increasing sensitivity to mating pheromone. As a result of these changes, somatostatin induced a dose-dependent increase in growth on media lacking histidine. Thus, the strain provides a sensitive bioassay to examine the molecular nature of interactions between the ligand and receptor and between the receptor and its cognate heterotrimeric G-protein complex. Moreover, the system should be generally adaptable for the discovery of novel, therapeutically active ligands for receptors of medical importance.

Figure 1 Somatostatin-dependent growth of yeast. (a) alpha-thinspace or a-factor binds to a seven-transmembrane domain receptor (STE2/3). As a result, the heterotrimeric G-protein complex dissociates relieving the inhibition of the Galpha subunit on the Gbetagamma dimer. The Galpha subunit interacts with the receptor protein at its carboxyl terminus and with the Gbetagamma dimer at its amino terminus. Once dissociated from Galpha, the Gbetagamma dimer then stimulates an MAPK module (recently reviewed in [33]), which has two consequences. Firstly, it stimulates FAR1, which in turn causes cell cycle arrest by interfering with G1 cyclin function. Secondly, it activates the STE12 transcription factor, which activates FUS3 as well as a diverse array of genes whose action is required for karyogamy. (b) The mammalian SSTR2 receptor is expressed in a his3 sst1 DeltaFUS1 strain together with a modified Galpha subunit (Galphai2), which interacts with SSTR2 at its carboxyl terminus and the yeast Gbetagamma proteins at its amino terminus. When somatostatin binds to SSTR2, Galphai2 dissociates from the heterotrimeric G-protein complex and Gbetagamma stimulates the MAPK module. Because the FUS1 has been deleted, cell cycle arrest does not occur. Stimulation of STE12 results in the activation of HIS3 fused to the FUS3 upstream sequences targeted by STE12. Expression of HIS3 allows growth on histidine–medium. The sst1 allele renders the cell hypersensitive to alpha-factor and consequently increases the sensitivity of the engineered cell to somatostatin.

Return to text reference [1]

Receptor-type tyrosine kinases, such as Src and Abl, play a key role in regulating cell proliferation and other processes in mammalian cells. They form part of the signal transduction pathway. Upon ligand binding to the extracellular domain, the receptors dimerize, resulting in autophosphorylation and consequent activation. The Src family of receptor kinases are negatively regulated by a discrete group of tyrosine kinases called the Csk family. These phosphorylate members of the Src family at a conserved tyrosine (Tyr527). This maintains the receptors in a low level of activity in the absence of ligand binding. Tyrosine kinases present an attractive target for small drug action to regulate cell proliferation. Two papers describe yeast-based screens for regulators and antagonists of their action [20••] [21••].

Expression of the Src tyrosine kinase is lethal in S. pombe, so cells expressing Src from a repressible thiamine promoter are unable to grow in the absence of thiamine. When such cells are transformed with a cDNA library, derived from either SV40 large T transformed human lung fibroblasts or human Burkitt lymphoma cells, only clones expressing negative regulators of Src are able to grow in the absence of thiamine. This screen isolated two classes of regulators [21••]. The first corresponded to the Csk family of Src regulators. The second were tyrosine phosphatases that inactivated Src itself or dephosphorylated its targets. Although both classes of regulators were already known, the results clearly validate the methodology. It is possible that these regulators are over-represented in the cDNA libraries used. Alternatively, there may be no more regulators left to identify.

Further members of the Src family can be isolated by screens based on conserved sequences. However, there is little sequence conservation in the ligands to which they bind. A screen in S. cerevisiae to identify such ligands has been developed. There are only very small amounts of proteins phosphorylated on tyrosine residues in a normal yeast cell. However, heterologous expression of an Src-family kinase leads to an increase that is readily detected by antiphosphotyrosine antibodies. The level is dramatically increased by the simultaneous expression of a known ligand. A yeast cell expressing the fibroblast growth factor (FGF) receptor gene was transformed with a Xenopus cDNA library and screened by colony immunoblotting. Six novel sequences were recovered. Of particular interest were two genes, FRL1 and FRL2, which are distantly related to epidermal growth factor (EGF) and angiotensin respectively. Both genes activated the FGF receptors in Xenopus oocytes. Overexpression of both genes induces mesoderm and neural specific genes in explants, and they are expressed at different stages during development. FRL1 is broadly expressed during gastrulation and neuralation; FRL2 is expressed principally in the axial mesoderm and brain at later stages.



Multidrug resistance proteins

The overexpression of P-glycoproteins in human cancers has been shown to confer resistance to a variety of unrelated chemotherapeutic drugs. Such proteins are members of the ATP-binding cassette (ABC) superfamily of membrane transporters. Other members of this group include the yeast a-mating pheromone transporter Ste6, and the human cystic fibrosis transmembrane conductance regulator (CFTR) protein, mutations of which result in cystic fibrosis. Expression of human multidrug resistance associated protein (MRP), or the mouse MRP Mdr3, in an anthracycline supersensitive yeast restored resistance to adriamycin, and conferred resistance to the unrelated antifungal agents FK506 and valinomycin [22••]. Radiolabelling experiments suggested that adriamycin was removed by an active transport mechanism. Human MRP also partially complements a sterile ste6 allele, suggesting that although MRP is only weakly homologous in structure to Ste6p, it can mediate the transport of the a-mating pheromone. Yeast may therefore prove to be extremely useful for the biochemical and pharmacological characterization of eukaryote ABC transporters.



Cytochrome P450

Cytochrome P450 and NADPH–cytochrome P450 oxidoreductase (OR) are key enzymes in the metabolism of xenobiotics, therapeutically active drugs and carcinogens. The heterologous expression of human P450s in yeast provides a valuable model system for their study. The expression of human P450s in yeast is limited by the activity of yeast OR (yOR) and the availability of internal membranes in which they are anchored. The lack of yOR may be overcome by the fusion of CYPIAI, the human gene encoding P450AI, to human OR (hOR) and the expression of the chimeric protein in yeast [23••]. This results in the expression of a catalytically self-sufficient, hybrid protein correctly located in internal membranes. The quantity of internal membranes may be increased by the overproduction of chimeric proteins consisting of CYPIAI fused to HMG2/1, which comprises the seven transmembrane domains of the S. cerevisiae 3-hydroxy-3-methylglutaryl-CoA reductase isozymes 1 and 2 [24••]. The hybrid protein was catalytically active and correctly located in the internal membranes. Replacement of the amino-terminal membrane anchor domain of human NADPH–cytochrome P450 OR by the HMG2/1 peptide also resulted in a functional fusion enzyme that was able to interact with the HMG2/1–CYPIAI fusion and the yeast endogenous P450 enzyme lanosterol-14-demethylase.



Technical advances


Co-expression of proteins that aid folding and secretion

There are many examples in which the expression of proteins in yeast is apparently limited by bottlenecks in the secretory pathway. Two recent papers [25] [26] suggest ways in which this may be overcome. The secretion of human leukocyte protease inhibitor was found to be elevated three-thinspace to fourfold by simultaneous overexpression of ubiquitin from a chromosomal UBI4 gene under the control of the GAL1 promoter [25]. Similarly, the overexpression of protein disulphide isomerase resulted in a 10-fold increase in the levels of secretion of human platelet-derived growth factor and a fourfold increase in the levels of acid phosphatase [26].



Plasmids to control the level of overexpression

The expression of proteins in yeast is often undertaken for reasons of fundamental research. Many investigations require the ectopic expression of a protein under the control of promoters programming different levels of expression. A convenient set of vectors has been developed that allows the constitutive level of proteins to be expressed over a range of three orders of magnitude [27] [28]. The gene to be expressed is cloned into a multiple cloning site situated between the promoter and the CYC1 transcriptional terminator. The promoter may be the weak CYC1 promoter, the ADH promoter or the stronger TEF1 and GPD promoters. The set is based on centromeric or 2 µm vectors.



Copy number and stability of integrated constructs in H. polymorpha

Because stable replicating vectors are not available for methylotrophic yeasts, genes must be chromosomally integrated. This can make it difficult to obtain strains harbouring a high copy number of the heterologous gene. In leu2 S. cerevisiae strains, the copy number of plasmids carrying the leu2d allele is elevated compared to plasmids bearing a wild-type LEU2 sequence. This is thought to be due to the inefficient expression of Leu2 leading to selection for cells with more copies of the plasmid. A similar process can be used to increase the copy number of chromosomally integrated genes in H. polymorpha [29•]. When a ura3 strain is transformed with a plasmid carrying the S. cerevisiae URA3 gene, the plasmid replicates in a highly unstable fashion. Presumably, this is due to a fortuitous ARS sequence in the URA3 gene. After prolonged growth in nonselective conditions, the replicating form of the plasmid disappears and is replaced by multiple copies (5–30 per cell), integrated head-to-tail in tandem arrays at a chromosomal site that is not the homologous URA3 locus. The high copy number probably reflects inefficient expression of the heterologous URA3 gene, leading to selection for increased copy number. It is not clear whether a single copy of the plasmid integrates first, followed by integration events of further plasmids at the same site, or whether multimerization of the plasmid occurs prior to a single integration event.

A similar phenomenon was observed during the expression of recombinant haemoglobin (rHbA) in H. polymorpha [30], using the S. cerevisiae LEU2 gene for selection. The construct carrying the alpha and beta chains was chromosomally integrated. After 40 generations in a chemostat, amplification was observed. The pressure for amplification is presumed to arise from the inefficient expression or function of the heterologous LEU2 gene. In some cases, the LEU2 gene was amplified but the globin expression cassettes were lost. In other cases, the expression cassettes were also amplified and there was a corresponding increase in the yield of rHbA. Such isolates maintained the increase when grown in fed-batch fermentation, producing fully functional rHbA.

Selection based on an integrated copy of the homologous H. polymorpha LEU2 gene can also be unstable [31•]. Despite vectors lacking any ARS sequence, unstable replicating plasmids arise that can retransform E. coli. Apparently, rearrangements result in the acquisition of a chromosomal ARS sequence. The same group have also described an elegant procedure for the targeted integration of an expression cassette to the MOX locus [32•]. It relies upon the TRP3 gene, which lies immediately downstream of the MOX locus. A strain is generated in which the TRP3 gene is disrupted by insertion/deletion with the S. cerevisiae LEU2 gene. The expression cassette is constructed to carry 5' MOX promoter sequences at one end and the TRP3 sequences at the other, extending beyond the sequences replaced by the LEU2 gene. Transplacement of the disrupted locus with the expression cassette thus regenerates a Trp+ strain that can readily be selected.



Conclusions

Where the expression of recombinant protein is the only objective, methylotrophic yeasts are now the preferred option. There are now many examples in which proteins have been expressed at significantly higher yields in methylotrophic yeasts, or even in which the successful expression in a methylotrophic yeast of proteins that completely failed to be expressed in S. cerevisiae has been achieved. Apart from the advantages discussed above, a well-designed commercial kit based upon P. pastoris is now available from InVitrogen. This should facilitate the use of such systems for those new to the field. The only note of caution is that there is a long history of the safe use of products derived from S. cerevisiae. The relative newness of methylotrophic systems may make the acceptance of recombinant products by regulatory authorities more difficult. Where expression of a recombinant protein is to be the basis of a model system to be used to develop other products, for example, in a drug screen, the exquisite genetic system of S. cerevisiae is likely to ensure that it will remain the predominant system for the foreseeable future.



References and recommended reading
Papers of particular interest, published within the annual period of review, have been highlighted as:
• of special interest
•• of outstanding interest
  1. Romanos MA, Scorer CA, Clare JJ:
    Foreign gene expression in yeast.
    Yeast 1992, 8: 423–488. [MEDLINE] [Cited by]
    Return to citation reference [1]

  2. Goodey AR:
    The production of heterologous plasma proteins.
    Trends Biotechnol 1993, 11: 430–433. [MEDLINE] [Cited by]
    Return to citation reference [1]

  3. Cregg JM, Vedvick TS, Raschke WC:
    Recent advances in the expression of foreign genes in Pichia pastoris.
    Bio-Technology 1993, 11: 905–909.
    Return to citation reference [1] [2]

  4. • Faber KN, Harder W, Ab G, Veenhuis M:
    Methylotrophic yeasts as factories for the production of foreign proteins.
    Yeast 1995, 11: 1331–1344. [MEDLINE] [Cited by]
    This paper (see also [3]) reviews recent developments in the production of heterologous proteins in methylotrophic yeast.
    Return to citation reference [1]

  5. Laroche Y, Storme V, De Meutter J, Messens J, Lauwereys M:
    High-level secretion and very efficient isotopic labeling of tick anticoagulant peptide (TAP) expressed in the methylotrophic yeast, Pichia pastoris.
    Bio-Technology 1994, 12: 1119–1124.
    Return to citation reference [1]

  6. Choi ES, Sohn JH, Rhee SK:
    Optimization of the expression system using galactose-inducible promoter for the production of anticoagulant hirudin in Saccharomyces cerevisiae.
    Appl Microbiol Biotechnol 1994, 42: 587–594. [MEDLINE] [Cited by]
    Return to citation reference [1]

  7. Heim J, Takabayashi K, Meyhack B, Marki W, Pohlig G:
    C-terminal proteolytic degradation of recombinant desulfato-hirudin and its mutants in the yeast Saccharomyces cerevisiae.
    Eur J Biochem 1994, 226: 341–353. [MEDLINE] [Cited by]
    Return to citation reference [1]

  8. • Hottiger T, Kuhla J, Pohlig G, Fuerst P, Spielmann A, Garn M, Haemmerli S, Heim J:
    2-mu-vectors containing the Saccharomyces cerevisiae metallothionein gene as a selectable marker: excellent stability in complex media, and high-level expression of a recombinant protein from a CUP1-promoter-controlled expression cassette in cis.
    Yeast 1995, 11: 1–14. [MEDLINE] [Cited by]
    Describes a selection for plasmid maintenance in complex medium. It cleverly links the intensity of the selective pressure to the degree of expression of the recombinant protein.
    Return to citation reference [1] [2]

  9. • Kim CH, Sohn JH, Choi ES, Rhee SK:
    Effect of soybean oil on the enhanced expression of hirudin gene in Hansenula polymorpha.
    Biotechnol Lett 1996, 18: 417–422. [Cited by]
    An unexpected and very original observation that the addition of soybean oil increases the yield of recombinant hirudin in H. polymorpha.
    Return to citation reference [1] [2]

  10. Lee DH, Park JB, Seo JH, Choi ES, Lee SK:
    Expression of hirudin in fed-batch cultures of recombinant Saccharomyces cerevisiae.
    Biotechnol Lett 1994, 16: 667–670. [Cited by]
    Return to citation reference [1]

  11. Mendozavega O, Hebert C, Brown SW:
    Production of recombinant hirudin by high cell density fed-batch cultivations of a Saccharomyces cerevisiae strain: physiological considerations during the bioprocess design.
    J Biotechnol 1994, 32: 249–259. [MEDLINE] [Cited by]
    Return to citation reference [1]

  12. Park JB, Kweon YE, Rhee SK, Seo JH:
    Production of hirudin by recombinant Saccharomyces cerevisiae in a membrane-recycle fermentor.
    Biotechnol Lett 1995, 17: 1031–1036. [Cited by]
    Return to citation reference [1]

  13. Weydemann U, Keup P, Piontek M, Strasser AWM, Schweden J, Gellissen G, Janowicz ZA:
    High-level secretion of hirudin by Hansenula polymorphathinspace authentic processing of three different preprohirudins.
    Appl Microbiol Biotechnol 1995, 44: 377–385. [MEDLINE] [Cited by]
    Return to citation reference [1] [2]

  14. •• Ridder R, Schmitz R, Legay F, Gram H:
    Generation of rabbit monoclonal antibody fragments from a combinatorial phage display library and their production in the yeast Pichia pastoris.
    Bio-Technology 1995, 13: 255–260.
    The efficient expression of antibody fragments in P. pastoris is a major advance. This is a major area in biotechnology. Previous attempts in S. cerevisiae have been disappointing.
    Return to citation reference [1]

  15. Vozza LA, Wittwer L, Higgins DR, Purcell TJ, Bergseid M, Collins RLA, Lavallie ER, Hoeffler JP:
    Production of a recombinant bovine enterokinase catalytic subunit in the methylotrophic yeast Pichia pastoris.
    Bio-Technology 1996, 14: 77–81.
    Return to citation reference [1]

  16. Rodriguez M, Rubiera R, Penichet M, Montesinos R, Cremata J, Falcon V, Sanchez G, Bringas R, Cordoves C, Valdes M, Lleonart R, Herrera L, De La Fuente J:
    High level expression of the b. microplus bm86 antigen in the yeast Pichia pastoris forming highly immunogenic particles for cattle.
    J Biotechnol 1994, 33: 135–146. [MEDLINE] [Cited by]
    Return to citation reference [1] [2]

  17. Kaslow DC, Bathurst IC, Lensen T, Ponnudurai T, Barr PJ, Keister DB:
    Saccharomyces cerevisiae recombinant pfs25 adsorbed to alum elicits antibodies that block transmission of Plasmodium falciparum.
    Infect Immun 1994, 62: 5576–5580. [MEDLINE] [Cited by]
    Return to citation reference [1]

  18. •• Faber KN, Westra S, Waterham HR, Keizergunnink I:
    Foreign gene expression in Hansenula polymorpha. A system for the synthesis of small functional peptides.
    Appl Microbiol Biotechnol 1996, 45: 72–79. [MEDLINE] [Cited by]
    Two important developments are reported in this paper: firstly, the targeting of a recombinant protein to peroxisomes, and secondly, fusion to a carrier protein to stabilize small peptides.
    Return to citation reference [1]

  19. •• Price LA, Kajkowski EM, Hadcock JR, Ozenberger BA, Pausch MH:
    Functional coupling of a mammalian somatostatin receptor to the yeast pheromone response pathway.
    Mol Cell Biol 1995, 15: 6188–6195. [Full text] [MEDLINE] [Cited by]
    See annotation [21••].
    Return to citation reference [1] [2]

  20. •• Superati-Furga G, Jonsson K, Courtneidge SA:
    A functional screen in yeast for regulators and antagonizers of heterologous protein tyrosine kinases.
    Nat Biotechnol 1996, 14: 600–605. [MEDLINE] [Cited by]
    See annotation [21••].
    Return to citation reference [1] [2]

  21. •• Kinoshita N, Minshull J, Kirschner MW:
    The identification of two novel ligands for FGF receptor by a yeast screening method and their activity in Xenopus development.
    Cell 1995, 83: 621–630. [MEDLINE] [Cited by]
    These three papers [19••] [20••] [21••] fully exploit the power of yeast genetics to establish functional models of important cell regulators.
    Return to citation reference [1] [2] [3] [4] [5]

  22. •• Ruetz S, Brault M, Kast C, Hemenway C:
    Functional expression of the multidrug resistance-associated protein in the yeast Saccharomyces cerevisiae.
    J Biol Chem 1996, 271: 4154–4160. [Full text] [MEDLINE] [Cited by]
    This paper establishes S. cerevisiae as a model system to study a medically important area.
    Return to citation reference [1]

  23. •• Wittekindt NE, Wurgler FE, Sengstag C:
    Functional expression of fused enzymes between human cytochrome p4501a1 and human NADPH–cytochrome p450 oxidoreductase in Saccharomyces cerevisiae.
    DNA Cell Biol 1995, 14: 273–283. [MEDLINE] [Cited by]
    This paper, together with [24••], describes elegant manipulation of S. cerevisiae to increase expression of human cytochrome P450s.
    Return to citation reference [1] [2]

  24. •• Wittekindt NE, Wurgler FE, Sengstag C:
    Targeting of heterologous membrane proteins into proliferated internal membranes in Saccharomyces cerevisiae.
    Yeast 1995, 11: 913–928. [MEDLINE] [Cited by]
    See annotation [23••].
    Return to citation reference [1] [2]

  25. Chen Y, Pioli D, Piper PW:
    Overexpression of the gene for polyubiquitin in yeast confers increased secretion of a human leucocyte protease inhibitor.
    Bio-Technology 1994, 12: 819–823.
    Return to citation reference [1] [2]

  26. Robinson AS, Hines V, Wittrup KD:
    Protein disulphide isomerase overexpression increases secretion of foreign proteins in Saccharomyces cerevisiae.
    Bio-Technology 1994, 12: 381–384.
    Return to citation reference [1] [2]

  27. Mumberg D, Muller R, Funk M:
    Yeast vectors for the controlled expression of heterologous proteins in different genetic backgrounds.
    Gene 1995, 156: 119–122. [MEDLINE] [Cited by]
    Return to citation reference [1]

  28. Mumberg D, Muller R, Funk M:
    Regulatable promoters of Saccharomyces cerevisiae: comparison of transcriptional activity and their use for heterologous expression.
    Nucleic Acids Res 1994, 22: 5767–5768. [MEDLINE] [Cited by]
    Return to citation reference [1]

  29. • Gatzke R, Weydemann U, Janowicz ZA, Hollenberg CP:
    Stable multicopy integration of vector sequences in Hansenula polymorpha.
    Appl Microbiol Biotechnol 1995, 43: 844–849. [MEDLINE] [Cited by]
    An important strategy for increasing the copy number of an integrated expression construct.
    Return to citation reference [1]

  30. Gilbert SC, Van UH, Greenfield AJ, McAvoy MJ, Denton KA, Coghlan D, Jones GD, Mead DJ:
    Increase in copy number of an integrated vector during continuous culture of Hansenula polymorpha expressing functional human haemoglobin.
    Yeast 1994, 10: 1569–1580. [MEDLINE] [Cited by]
    Return to citation reference [1]

  31. • Bogdanova AI, Agaphonov MO, Ter AMD:
    Plasmid reorganization during integrative transformation in Hansenula polymorpha.
    Yeast 1995, 11: 343–353. [MEDLINE] [Cited by]
    A cautionary tale for those who assume that integrated sequences are always stable. Probably results from the use of the homologous LEU2 gene.
    Return to citation reference [1]

  32. • Agaphonov MO, Beburov MY, Ter AMD, Smirnov VN:
    A disruption-replacement approach for the targeted integration of foreign genes in Hansenula polymorpha.
    Yeast 1995, 11: 1241–1247. [MEDLINE] [Cited by]
    An elegant method for forced integration of an expression cassette.
    Return to citation reference [1]

  33. Wittenberg C, Reed SI:
    Plugging it in: signalling circuits and the yeast cell cycle.
    Curr Opin Cell Biol 1996, 8: 223–230. [Full text] [MEDLINE] [Cited by]
    Return to citation reference [1]



Author Contacts
PE Sudbery, Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, S10 2TN, UK; e-mail: p.sudbery@sheffield.ac.uk.
Return to author list


Copyright

Copyright © 1996 Current-Opinion.com
Help   Feedback      Search   Map
Black Line
© 1998-1999 BioMedNet. All rights reserved.
email: info@biomednet.com.