Vous êtes sur la page 1sur 14

Metabolic Engineering xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Metabolic Engineering
journal homepage: www.elsevier.com/locate/meteng

Review

Metabolic engineering of Pichia pastoris


David A. Peñaa,b, Brigitte Gassera,b,c, Jürgen Zanghellinia,b, Matthias G. Steigera,b,

Diethard Mattanovicha,b,
a
Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
b
Austrian Centre of Industrial Biotechnology, Vienna, Austria
c
CD-Laboratory for Growth-Decoupled Protein Production in Yeast, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria

A R T I C LE I N FO A B S T R A C T

Keywords: Besides its use for efficient production of recombinant proteins the methylotrophic yeast Pichia pastoris (syn.
Pichia pastoris Komagataella spp.) has been increasingly employed as a platform to produce metabolites of varying origin. We
Komagataella summarize here the impressive methodological developments of the last years to model and analyze the me-
Metabolic engineering tabolism of P. pastoris, and to engineer its genome and metabolic pathways. Efficient methods to insert, modify
Synthetic biology
or delete genes via homologous recombination and CRISPR/Cas9, supported by modular cloning techniques,
Systems biology
Bioeconomy
have been reported. An outstanding early example of metabolic engineering in P. pastoris was the humanization
of protein glycosylation. More recently the cell metabolism was engineered also to enhance the productivity of
heterologous proteins. The last few years have seen an increased number of metabolic pathway design and
engineering in P. pastoris, mainly towards the production of complex (secondary) metabolites. In this review, we
discuss the potential role of P. pastoris as a platform for metabolic engineering, its strengths, and major re-
quirements for future developments of chassis strains based on synthetic biology principles.

1. Introduction Komagataella strains employed in biotechnological applications.


At a time when the genome of Saccharomyces cerevisiae was se-
The yeast commonly known as Pichia pastoris has first been de- quenced (Goffeau et al., 1996), laying foundations for metabolic en-
scribed as Zygosaccharomyces pastori by the French mycologist and cy- gineering in this yeast (Nielsen, 1998), nearly nothing was known of
tologist Alexandre Guilliermond (Guilliermond, 1920). In the 1950s, genomics, metabolism (except for methanol metabolism) and cell
Herman Phaff isolated several related strains from oak trees in Cali- biology of P. pastoris. It took until 2009 when two genome sequences
fornia, and renamed the species as Pichia pastoris (Phaff et al., 1956). were published (De Schutter et al., 2009; Mattanovich et al., 2009),
Such as only few other yeasts, it shows a distinct ability to grow on followed by further (re)-sequencing and re-annotation of several
methanol as the sole carbon and energy source, which is based on strains: both the K. pastoris type strain CBS704/DSMZ70382 and K.
several highly overexpressed genes encoding the enzymes of the me- phaffii CBS7435 and its commercial variant GS115 (Kuberl et al., 2011;
thanol assimilation and dissimilation pathways (Wegner and Harder, Love et al., 2016; Sturmberger et al., 2016; Valli et al., 2016). Thor-
1987). In the 1980s the feature to highly express genes such as AOX1 oughly annotated genome sequences are available at www.
(encoding for methanol oxidase, the first step of methanol utilization) pichiagenome.org and have formed the basis for detailed systems
only when methanol is present, was utilized to develop a strong and biology analyses of P. pastoris since then (Zahrl et al., 2017).
methanol inducible expression system (Cregg et al., 1985). In 1995 P. P. pastoris is a methylotrophic yeast, capable to oxidize methanol for
pastoris was re-classified into the newly established genus Komagataella energy production, and to assimilate it as sole carbon source for growth
(Yamada et al., 1995), which was later split into several species and product formation. After oxidation of methanol to formaldehyde,
(Kurtzman, 2005), so that former P. pastoris strains now split up into the assimilation begins with the reaction of dihydroxyacetone synthase, a
two species K. pastoris and K. phaffii. Today six species of the genus specialized transketolase, transferring formaldehyde to xylulose-5-
Komagataella are described (Naumov et al., 2013). Thus, the widely phosphate (Xu5P), resulting in glyceraldehyde-3-phosphate and dihy-
accepted name Pichia pastoris stands for at least two different species. droxyacetone. Xu5P is then regenerated via a cyclic pathway (called the
To avoid confusion and to include all strains used in biotechnology we Xu5P cycle) involving reactions of the pentose phosphate pathway.
use the established name P. pastoris here as a synonym for all Russmayer et al. (2015a) showed recently that the entire Xu5P cycle is


Corresponding author at: Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.
E-mail address: diethard.mattanovich@boku.ac.at (D. Mattanovich).

https://doi.org/10.1016/j.ymben.2018.04.017
Received 22 February 2018; Received in revised form 16 April 2018; Accepted 23 April 2018
1096-7176/ © 2018 Published by Elsevier Inc. on behalf of International Metabolic Engineering Society.

Please cite this article as: Peña Navarro, D.A., Metabolic Engineering (2018), https://doi.org/10.1016/j.ymben.2018.04.017
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

localized in peroxisomes, utilizing a set of specialized enzymes encoded 2.1. Quantification of cellular metabolism
by duplicated genes that acquired transcriptional regulation by me-
thanol. This finding sheds light on the potential benefit of compart- 2.1.1. Metabolomics
mentalization of an entire specialized pathway and may be utilized for The set of metabolites synthesized and taken up by an organism
metabolic pathway engineering in the future. constitute its metabolome (Fiehn, 2002). “The metabolome can be de-
Besides methanol, glucose and glycerol are frequently used as fined as the complete complement of all small molecule (< 1500 Da)
carbon sources. Glycerol is utilized quite fast (qSmax ca. 0.37 h−1 in metabolites found in a specific cell, organ or organism” (Wishart,
mineral media) with a high biomass yield making it a useful substrate to 2007). This definition by Wishart includes all small metabolites but
accumulate biomass, recombinant proteins and potentially metabolites. excludes polymers like DNA, RNA, proteins, polymeric carbohydrates
The presence of four putative H+/glycerol symporters encoded in the and others. Importantly, also metabolites detected and not necessarily
genome is an explanation for the efficient utilization of glycerol synthesized by the organism are included in this definition. Metabo-
(Mattanovich et al., 2009). lomics is the technology used to measure and describe qualitatively and
Glucose uptake is limited in P. pastoris compared to S. cerevisiae, quantitatively the metabolic state of a cell. It directly provides in-
with a nearly tenfold difference in maximum specific glucose uptake formation about alterations in the cellular metabolism and can reveal
rates (qSmax ≈ 0.35 h−1 and 2.88 h−1, respectively). Under fully potential targets for metabolic engineering. However, compared to
aerobic conditions the glycolytic flux does not exceed the respiratory other omics technologies like transcriptomics or genomics, metabo-
capacity, so that nearly no fermentative by-products are formed lomics has the challenge that the chemical diversity of metabolites is
(Hagman et al., 2014). P. pastoris is therefore classified as a canonical substantially larger. For genomics, transcriptomics and to a lesser ex-
Crabtree-negative yeast. This limitation of glucose uptake in compar- tent for proteomics, the analytes (DNA, RNA, proteins) are chemically
ison to S. cerevisiae has been ascribed to the low number of hexose homogenous, which facilitates the establishment of a general metho-
transporter genes and a potentially more rigid regulatory network of dology to quantify these entities.
carbon metabolism. The lower glycolytic flux comes with a higher For P. pastoris, a first benchmark study providing quantitative me-
fraction of pentose phosphate pathway (PPP) flux with a split ratio of tabolite data was published 2012 by Carnicer et al. (2012a). In this
40–50% PPP flux (Baumann et al., 2010; Nocon et al., 2016). This work more than 30 metabolites including amino acids, organic acids
provides much more NADPH regeneration per carbon compared to S. and some sugar phosphates were quantified after controlled cultivation
cerevisiae, which has a split ratio in glucose surplus conditions in the in a chemostat, using an optimized quenching protocol. Importantly, an
range of 4–17% (Gombert et al., 2001; Maaheimo et al., 2001; interspecies comparison between S. cerevisiae and P. pastoris was per-
Velagapudi et al., 2007). Apart from these fundamental differences in formed, highlighting some differences between the two species. While
glucose utilization it was shown recently that P. pastoris reduced its the metabolite concentrations of upper glycolysis and the tricarboxylic
maintenance demand threefold under extreme calorie restriction acid (TCA) cycle were comparable, the concentrations of metabolites of
(Rebnegger et al., 2016) different to S. cerevisiae where no change was the lower part of glycolysis (2-phosphoglycerate/3-phosphoglycerate
observed (Vos et al., 2016). and phosphoenolpyruvate pools) were lower in P. pastoris. Interestingly,
This review is intended as a comprehensive summary of the devel- also the pool of trehalose-6-phosphate was significantly larger in S.
opment and current state of the art of metabolic engineering in P. cerevisiae compared to P. pastoris (Carnicer et al., 2012a).
pastoris and the underlying specific techniques to analyze and engineer An important part in metabolomics is dealing with sample pre-
its metabolism. For comprehensive reviews on cell engineering meth- paration techniques due to two main reasons: first, as already men-
odology and process design, the reader is referred to reviews by tioned, chemically heterogeneous compounds need to be analyzed, and
Schwarzhans et al. (2017a) and Yang and Zhang (2018). The first one second, the cellular metabolism needs to be arrested rapidly to measure
focusses on metabolic and cell engineering, systems biology and an metabolites with a high turnover rate. In order to stop cellular meta-
extensive discussion of clonal variability. The latter addresses most bolism, cells are quenched and consecutively washed to remove ex-
specifically bioreactor cultivation design as a tool for optimization of tracellular metabolites. During this procedure it is important that cells
protein production. More specific reviews on P. pastoris engineering are not retained for a long period of time in the pure quenching solution
focused on glycoengineering (Laukens et al., 2015), mathematical as this leads to substantial leakage of metabolites to the quenching
modeling (Theron et al., 2018), systems biotechnology (Zahrl et al., solution. For P. pastoris cells, a rapid filtration and washing step on a
2017) and CRISPR-Cas technologies in different yeasts (Raschmanová filter is best suited to remove extracellular metabolites and reduce
et al., 2018). unwanted leakage effects (Russmayer et al., 2015b). The initial
Based on conclusions from past achievements, novel developments quenching procedure established by (Carnicer et al., 2012a) for P.
will be discussed here in the light of recent synthetic biology progress, pastoris using 60% (v/v) methanol was further optimized by buffering
drawing conclusions for the upcoming potential of this yeast for the quenching solution to reduce the leakage of metabolites during the
modern metabolic engineering. quenching process. In order to provide a framework for further buffer
optimizations the parameters influencing the leakage of a certain me-
2. Methods of metabolic engineering of P. pastoris tabolite were investigated. Hereby, it was found that size and charge-
related properties of a metabolite play a major role in controlling me-
Until recently, specific methods to engineer and analyze the meta- tabolite loss, which are: molecular weight, the van-der-Waals volume,
bolism of P. pastoris have been quite underdeveloped, allowing only the charge-weighted positive surface area, charge-weighted negative
single solutions to specific questions rather than targeted construction surface area and the total polar surface area. Depending on the main
of chassis platforms for advanced metabolic engineering. Since the research question and the metabolites of interest, these influence fac-
publication of the first genome sequences in 2009 this development has tors can help to decide which buffer systems are best suited. The buffer
gained great momentum. This section summarizes the current state of system that has shown so far the best properties towards reducing
the art of methods to quantify intracellular metabolites and metabolic leakage losses is Tris, buffered (0.125 M) to a pH of 8.2, with a NaCl
fluxes, to model and predict metabolic states, and to engineer cellular concentration of 0.055 M and a methanol concentration of 60% (v/v)
metabolism of P. pastoris for the production of new biomolecules or the (Mattanovich et al., 2017). To gain metabolites out of the quenched
enhanced productivity and quality of heterologous proteins. Based on biomass for their analysis, the extraction method of choice employs
the impressive methodological developments, enabling advanced boiling ethanol (Carnicer et al., 2012a). However, in another early
Design-Build-Test cycles for metabolic engineering, P. pastoris holds a study a direct comparison of boiling ethanol and repeated freeze-thaw
position at the forefront of synthetic biology. with methanol plus sonication showed that also the freeze-thaw

2
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

extraction method can be as efficient as the boiling ethanol procedure compartmentalization compared to S. cerevisiae. This information is
(Tredwell et al., 2011). especially important for flux analysis experiments as the leucine pre-
In order to accurately quantify the absolute concentrations of me- cursor alpha-isopropylmalate is synthesized from alpha-ketoisovalerate
tabolites, internal standards are used which are either labeled with 13C consuming cytosolic acetyl-CoA instead of mitochondrial acetyl-CoA
or 15N. 13C labeled standards are preferred for the obvious reason that (Förster et al., 2014). Current experiments show that also other amino
carbon is present in most cellular metabolites. As for many metabolites acid biosynthesis pathway enzymes have a different compartmentali-
such labeled standards are commercially not available, it is common zation in P. pastoris compared to S. cerevisiae. In particular, biosynthetic
practice to utilize a uniformly 13C- labeled cell extract (Lu et al., 2017). enzymes of the arginine and lysine pathways were found in the per-
The preparation of such a standard for P. pastoris and its application oxisome or cytosol rather than in mitochondria (own unpublished
during the quenching and extraction procedure was described by data). In a recent publication comparing S. cerevisiae and the methy-
Neubauer et al. (2012). lotrophic yeast Hansenula (Ogataea) polymorpha it was highlighted that
Metabolomic datasets can be used for different research applica- it is important to carefully evaluate compartmentalization constraints
tions. One aspect is to analyze the metabolite concentrations in strains in modeling approaches and not to extrapolate assumptions regarding
producing recombinant proteins compared to control strains (Jorda compartmentalization from other organisms without further evidence
et al., 2014). For example, analysis on protein producing strains sup- (Lehnen et al., 2017).
plemented with all amino acids showed that costly amino acids were In order to obtain absolute flux values, it is necessary to determine
preferentially taken up (Heyland et al., 2011), suggesting a regulatory the biomass composition of the cell, besides other important parameters
logic applied under the limited energy and redox availability caused by such as metabolite uptake and secretion rates. In 2009, the macro-
recombinant protein production. Another research question was to molecular composition of P. pastoris cells under different oxygenation
characterize the metabolite levels on different carbon sources, espe- conditions was investigated and compared to a strain producing a re-
cially on methanol for P. pastoris, compared to glucose or glycerol combinant protein. It was found that the amino acid composition of P.
conditions. Connected to this aspect, a quantitative analysis of in- pastoris is significantly different from the amino acid composition de-
tracellular free metabolites has been used to study the metabolic impact termined for the yeast S. cerevisiae. Furthermore, it was found that
of assimilation of methanol as a carbon source (Russmayer et al., different oxygenation levels have an impact on the general biomass
2015a). Under methanol assimilation conditions, also the special me- composition and need to be taken into account (Carnicer et al., 2009).
tabolite sedoheptulose-1,7-bisphosphate was detected, which is re- The determined biomass datasets were integrated into a stoichiometric
quired for the xylulose-monophosphate cycle enabling methanol as- model together with flux ratios of a METAFoR analysis yielding abso-
similation. lute fluxes (Baumann et al., 2010). Biomass composition differs mark-
In many metabolomic studies, the main focus is on metabolites of edly in cells grown on different carbon sources as well, which should be
the central carbon and energy metabolism, which are typically com- taken into consideration for metabolic flux balancing (Tomàs-Gamisans
prising the metabolite classes of amino acids, organic acids, sugar et al., 2018).
phosphates and alcohols. However, a study investigating the physiolo- Different analytic techniques can be used to analyze labeling pat-
gical impacts of hypoxia also focused on the lipid metabolism and on terns of 13C labeling experiments. Due to the high abundance of pro-
the so called lipidome. Changes in lipid metabolites including fatty teinogenic amino acids, NMR (Solà et al., 2004) as well as GC-MS based
acids, phospholipids and sphingolipids were quantified under normoxic techniques (Heyland et al., 2011) were used to determine the labeling
and hypoxic conditions showing significant differences (Adelantado profile in P. pastoris cultures. Later also techniques were established to
et al., 2017). directly measure the labeling pattern of free intracellular metabolites
Most studies conducted with P. pastoris measuring intracellular based on GC-MS (Mairinger et al., 2015; Mairinger et al., 2018) and LC-
metabolites levels were targeted approaches. In contrast, Tredwell et al. MS (Jorda et al., 2013).
(2017) followed an untargeted strategy using NMR to evaluate the 13
C flux measurements were often used to determine the flux
impact of recombinant protein production on metabolomic profiles. changes between a protein producing strain compared to a control
Thereby, the untargeted NMR metabolic profiling strategy was com- strain (Table 1). In several studies, the flux profile in protein producing
bined with a transcript analysis of unfolded protein response (UPR) P. pastoris was shown to change towards an up-regulation of the PPP
relevant gene transcripts (HAC1, KAR2, PDI1). Correlations between pathway. Consistently, the engineering of the PPP pathway by over-
these UPR markers and certain metabolite signals (isoleucine, aspartate, expression of the first two enzymatic steps (encoded by ZWF1 and
arabitol) were detected assuming that such data can be used as in- SOL3) further increased recombinant protein production as discussed in
dicators to detect UPR stress. The benefit of replacing transcript ana- Section 3.2 (Nocon et al., 2016).
lysis of one or a few genes with a metabolomics method remains
however to be proven. 2.2. Metabolic modeling

2.1.2. Metabolic flux analysis Various (ad hoc) modeling efforts, very recently reviewed by Theron
13
C metabolic flux analysis is a powerful tool to experimentally et al. (2018), have been undertaken to understand and optimize espe-
determine the flux distribution in a cell (Sauer, 2006; Zamboni et al., cially protein production in P. pastoris. Here we outline recent devel-
2009). The methodology is used on a frequent basis to measure the opments in the modeling and genome-wide analysis of P. pastoris me-
metabolic flux distributions in P. pastoris, and Table 1 gives an overview tabolism. Constraint-based analysis is a key approach in the systems
about 13C metabolic flux experiments carried out with this organism. analysis of metabolism and has proven extremely useful in providing
In order to calculate fluxes, it was first necessary to verify which mechanistic insights into metabolism. Together with (genome-scale)
reactions occur in the central carbon metabolism of P. pastoris. As metabolic models, constraint-based analysis enables the prediction of
mentioned earlier, genome data for P. pastoris was not available until intracellular steady-state flux distributions and the study of the geno-
2009. Therefore, the first labeling experiments were also used to type-phenotype relations in silico (Lewis et al., 2012).
identify the biochemical pathways and to investigate if they differ from For P. pastoris several genome-scale metabolic models are currently
S. cerevisiae. It was found that the proteinogenic amino acids in P. available (Caspeta et al., 2012; Chung et al., 2010; Saitua et al., 2017;
pastoris are in principle synthesized as in S. cerevisiae and that the re- Sohn et al., 2010; Tomas-Gamisans et al., 2016; Tomàs-Gamisans et al.,
solved flux ratios of P. pastoris are more similar compared to S. cerevisiae 2018; Ye et al., 2017). The latest two genome-scale metabolic models
than to Scheffersomyces (Pichia) stipitis (Solà et al., 2004). However, represent two independent consensus reconstructions that integrated
later it was found that the leucine pathway has a different and updated the knowledge of the previous reconstructions. While

3
D.A. Peña et al.

Table 1
13
Main studies applying C metabolic flux analysis to P. pastoris.
13
Research question related to C labeling experiment Labeled substrate Measured metabolites Analytical procedure Ref.

Verification of amino acid biosynthesis pathways and flux ratio analysis after growth 10% (w/w) of [U-13C6] glucose or [U-13C3] glycerol Proteinogenic amino acids NMR (2D [13C,1H]- (Solà et al., 2004)
on either glucose and glycerol COSY)
Analysis and regulation of the methanol metabolism in the presence of a second carbon different mixtures of glycerol and methanol, always 10% (w/w) Proteinogenic amino acids NMR (2D [13C,1H]- (Solà et al., 2007)
source of [U-13C3] glycerol and [U-13C1] methanol COSY)
Flux changes between a protein producing strain and a control strain in fed-batch 10% (n/n) [U-13C6] glucose Proteinogenic amino acids GC-MS (Heyland et al., 2010)
experiments
13
Amino acid supplementation to unburden cellular metabolism during protein 10% (n/n) [U-13C6] C- glucose Proteinogenic amino acids GC-MS (Heyland et al., 2011)
production
The metabolic flux distribution during growth on a mixed feed of glucose and 12% (w/w) [U-13C6] glucose and [U-13C1] methanol Proteinogenic amino acids NMR (1H-13C-HSQC) (Jordà et al., 2012)
methanol while producing recombinant protein

4
13 13
Mapping of metabolic fluxes of glycolysis, pentose phosphate and methanol 80% [1– C1] glucose and 20% [U- C6] glucose; 100% Intracellular metabolites GC-MS, LC-MS (Jorda et al., 2013)
assimilation pathways [U-13C1] methanol
Comparison of fluxes between recombinant protein producing strain (Rol) and control 80% [1–13C1] glucose and 20% [U-13C6] glucose; 100% Intracellular metabolites GC-MS, LC-MS (Jorda et al., 2014)
strain in chemostat [U-13C1] methanol
Comparison of fluxes between high expression recombinant protein producing strain 80% [U-13C] glucose, 20% [1–13C] glucose Intracellular free amino GC-MS (Nie et al., 2014)
(beta-galactosidase) and low expression control strain acids
Comparison of fluxes between recombinant protein producing strain (hSOD) and 17% [U-13C6] glucose; Proteinogenic amino acids GC-MS (Nocon et al., 2014)
control strain
13 13
Flux distribution in chemostat cultivations of P. pastoris comparing glucose to a mixed 20% [U- C6] glucose; 20% [U- C1] methanol and 20% Proteinogenic amino acids GC-MS (Russmayer et al., 2015a)
substrate of methanol/glycerol [U-13C3] glycerol;
Measurement of split ratios between glycolysis and pentose phosphate pathway (PPP) 100% [1,6–13C; 2,3,4,5–12C] glucose; Intracellular metabolites GC-MS (Nocon et al., 2016)
analyzing strains engineered in the PPP
Differences in fluxes comparing medium with and without glutamate 80% [U-13C] glucose, 20% [1–13C] glucose Intracellular free amino GC-MS (P. Liu et al., 2016)
acids
13
High anabolic use of the TCA cycle on glucose and comparison of flux profile to S. 10% (w/w) U- C labeled Proteinogenic amino acids NMR (Zhang et al., 2017)
cerevisiae and P. stipitis
Metabolic Engineering xxx (xxxx) xxx–xxx
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

Tomàs-Gamisans et al. (2018) focused on extending the phenotypic synthesis steps, and/or absolute protein abundances – data that is not
capabilities by providing accurately measured biomass compositions readily available for non-classical model organisms.
during growth on glucose, glycerol and methanol as sole carbon source,
Ye et al. (2017) put their focus on increasing the coverage of their 2.3. Tools for metabolic engineering and synthetic biology of P. pastoris
model, which currently contains 1243 annotated genes, 2407 reactions,
and 1740 metabolites. In comparison, the latest version of the con- 2.3.1. Genomic integration: prerequisite for genetic engineering
sensus genome-scale reconstruction of S. cerevisiae, YEAST v7.0 (Aung Applying metabolic and cell engineering strategies requires ad-
et al., 2013) contains 910 genes, 3498 reactions and 2384 metabolites. vanced tools to perform genetic modifications and to introduce syn-
These recent model expansions led to an improved predictability of P. thetic pathways into the host (Wagner and Alper, 2016). Since the early
pastoris growth capabilities over a wide range of different carbon and days of P. pastoris research, gene overexpression has mostly been per-
nitrogen sources in industrially relevant conditions. Overall, current formed by stably introducing the gene expression cassette into the
genome-scale metabolic models of P. pastoris allow one to robustly genome, ideally into a targeted locus via a single crossover knock-in
predict cellular growth rates. In fact, these models were already used to strategy based on homologous recombination (HR). Although HR effi-
guide rational metabolic engineering projects, e.g., to improve re- ciencies are lower in comparison to S. cerevisiae, the number of trans-
combinant protein production (Nocon et al., 2014; see Section 3.2 for formants obtained is usually sufficient to select strains for protein
details), or to support culture media optimization (Matthews et al., production. Using the standard electroporation-based transformation
2018). To improve the predictive quality of these models not only with protocol described in Gasser et al. (2013), adapted from Cregg and
respect to growth but also with respect to recombinant protein yield, Russell (1998), or the high-efficiency protocol applying pretreatment
Irani et al. (2016) attempted to reconstruct the native as well as the with lithium acetate prior to electroporation (Wu and Letchworth,
humanized N-glycosylation pathways in P. pastoris. Although including 2004), transformation efficiencies of 103–104 colonies per µg linearized
N-glycosylation into the analysis reduced the predicted protein yield DNA are typically obtained, with around 85% of the obtained trans-
(compared to a purely metabolic analysis), the model typically over- formants containing the expression cassette in single or multiple copies.
estimated achievable protein yields by orders of magnitude. Thus, fu- Methods to increase multi-copy transformants which are often asso-
ture development will need to take other processes (e.g., protein folding ciated with higher gene expression and product levels have been re-
and secretion efficiencies) into account in order to realistically model cently reviewed by Piva et al. (2017).
protein production. Also the generation of gene knockouts was described early on, e.g.
Constraint-based modeling provides means to analyze the metabolic for protease-deficient mutants (Gleeson et al., 1998) or AOX1/2-defi-
steady-state behavior determined by the stoichiometric constraints of cient strains (Cregg et al., 1989), however, targeting efficiencies have
the biochemical reaction network, while regulatory constraints are often been a bottleneck for this purpose. While in principle also the
often not considered. In the case of P. pastoris the situation is ag- knock-in approach can be used to disrupt gene functions (Vervecken
gravated by the fact that knowledge on metabolic regulation specific to et al., 2004) in most cases generation of knock-outs relies on gene re-
P. pastoris is all but known. Computational methods that aim to com- placement by double crossover (Da Silva and Srikrishnan, 2012). De-
pensate these deficiencies by the integration of multi-omics data sets pending on the genomic locus to be disrupted and the length of
have so far proven unsuccessful (Machado and Herrgård, 2014). homologous flanking regions, gene replacement efficiencies can range
Moreover, recent findings by (Hackett et al., 2016) questioned the from < 0.1–80% (Nett and Gerngross, 2003; Schwarzhans et al., 2016;
importance of transcriptional regulation on changes in nutrient condi- Vogl et al., 2018a), and often a high number of false positive trans-
tions at least in S. cerevisiae. They report that many changes in flux formants was observed. For obvious reasons genes are easily dis-
levels are due to changes in the metabolome (by Michaelis-Menten-like ruptable when their deletion has no negative impact on strain fitness
kinetics) rather than changes in the enzyme levels. These observations while the disruption of genes with a negative impact on growth leads to
motivate the increased interest in the development of large-scale kinetic much lower frequencies of positive clones. In the latter case positive
models of metabolism (Vasilakou et al., 2016). clones face a negative selection pressure during cultivation so that
A (quasi-)dynamic analysis of P. pastoris was recently presented by clones with incorrect integration into random loci may dominate. As
Saitua et al. (2017). It coupled the uptake and secretion dynamics of long as sufficiently long homologous arms (approximately 1000 bps on
key metabolites with the steady-state behavior of metabolism in a well- each side) are provided, the probability of obtaining correctly in-
established framework known as dynamic flux-balance analysis tegrated clones is still rather high, however, if short or no flanking
(Höffner et al., 2013; Sánchez et al., 2014). By doing so, the authors regions are presented non-homologous end joining (NHEJ) is prevailing
could analyze the dynamic rearrangements in the intracellular flux (Näätsaari et al., 2012; own unpublished data). Deletion of the NHEJ
distributions. Moreover, they derived optimized feeding strategies and machinery allowed to use significantly shorter homologous flanking
beneficial single gene deletion strategies that led to a predicted increase regions (down to 250 bps on each side). However, the downside of this
in production of recombinant human serum albumin. However, the approach is that a specific genetic background bearing the Δku70 mu-
experimental viability of the predictions and the computational scal- tation needs to be used, which shows 10–30% reduced growth rates,
ability of the method to predict more complex intervention strategies reduced transformation efficiencies and decreased stress tolerance due
remain to be further investigated. to impaired DNA damage repair (Näätsaari et al., 2012; Weninger et al.,
Next to dynamic constraints, (global) resource allocation constraints 2018).
have received much attention in recent years (Berkhout et al., 2013; In contrast, the split-marker approach (Heiss et al., 2013) and
Goelzer and Fromion, 2011; Molenaar et al., 2009; O'Brien and Palsson, genome editing by CRISPR/Cas9-mediated homology directed repair
2015). In essence, these methods try to take enzyme levels into account HDR (Gassler et al., In press; Weninger et al., 2018, 2016) improve or
and assign a “price” on the expression of each protein/enzyme. As total trigger HR, independent of the strain background. The latter has also
resources are limited, cells have to carefully adjust their resource al- the advantage of marker-free genetic manipulations. Alternatively, re-
location, which – in turn – allows one to calculate optimal enzyme combinase-based marker recycling strategies such as Cre/loxP (Marx
distributions and corresponding flux distributions. Although im- et al., 2008; Pan et al., 2011) and FLP/FRT (Cregg and Madden, 1989;
pressively accurate results were achieved in Bacillus subtilis (Goelzer Näätsaari et al., 2012; Perez-Pinera et al., 2016) are available for P.
et al., 2015), Escherichia coli (O'Brien et al., 2013) and S. cerevisiae pastoris. While episomal plasmids are usually not in use for the ex-
(Sánchez et al., 2017) resource allocation has not been studied in P. pression of recombinant genes due to their segregational instability in
pastoris yet as these methods tend to be quite data intensive. They re- non-selective conditions (Hong et al., 2006; Liachko and Dunham,
quire i.a. estimations of catalytic rates, detailed knowledge of protein 2014; Sreekrishna et al., 1987), they proved to be an excellent choice

5
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

for genome engineering approaches such as the expression of Cas9 or expression units by assembly of standardized and commonly used ge-
Cre recombinase. Efficient curing of the episomal plasmid can be netic parts such as promoters, secretion signals, tags and transcription
achieved during one round of plating, meaning that the final production terminators (Obst et al., 2017 – MoClo Pichia Toolkit, available at
strain is free of the DNA modifying enzyme. Addgene #1000000108; Schreiber et al., 2017; Vogl et al., 2015). Ad-
ditionally, a library of nuclear localization signals was reported
2.3.2. Genome editing with CRISPR/Cas9 (Weninger et al., 2015).
In addition to gene replacements and targeted integration, CRISPR/ The GGA toolbox was then extended to allow simultaneous ex-
Cas9 can be used to create indel mutations via the NHEJ DNA repair pression of up to eight genes from one vector and the repertoire of
mechanism at targeted genomic loci if no homology target (also called synthetic parts encompasses 20 promoters, 10 transcription termina-
donor DNA) is provided. Frequencies are highly dependent on the tors, 5 integration loci and 4 resistance markers (Prielhofer et al., 2017)
genomic locus and the used guide RNAs, and range from 90% to 100% – GoldenPiCS kit, available at Addgene #1000000133. The GoldenPiCS
for easily disruptable genomic loci such as AOX1, DAS1/2 or the gly- kit is fully compatible with the GoldenMOCS system, which was de-
cerol kinase encoding gene GUT1, but can be as low as < 5% for dif- signed to perform metabolic engineering tasks in different cellular hosts
ficult loci and/or specific sgRNAs. It has been shown that in P. pastoris with the same cloning platform (Sarkari et al., 2017). Gibson assembly
CRISPR/Cas9 preferentially creates deletions of one single bp (and to a has also been used for the same purpose, with a set of 49 promoters and
lesser extent 2 or 3 bps) upstream of the Cas9 cleavage position, in- 20 terminators characterized (Vogl et al., 2016). Both studies found
dicating a difference to S. cerevisiae where insertions and deletions of that it is crucial to avoid repetitive sequences (such as the same pro-
different lengths (DiCarlo et al., 2013) were observed. moter or terminator) in vector or pathway design in order to prevent
Independently, both Weninger et al. (2016) and Gassler et al. (In loop out during the transformation step, which resulted in only partial
press) found that Cas9 exerts some toxicity to the host cells. Corre- integration of the desired pathway.
spondingly, the use of weaker promoters for Cas9 expression (such as Multiple enzyme pathways can be expressed using combinatorial
PPFK300 or PLAT1) was shown to enhance transformation efficiency and assembly of gene cassettes (by Gibson assembly or the GoldenPiCS
growth rate (Prielhofer et al., 2017). Furthermore, both groups strongly toolkit) or using single cassettes with polycistronic expression con-
recommend testing at least 2 or 3 different guide RNAs per locus. Also, structs based on 2 A peptide (Geier et al., 2015; Siripong et al., 2018),
the capacity for multiplexing (targeting two or more loci simulta- thus enabling efficient testing of complex heterologous pathways and
neously) has been demonstrated, with slightly reduced targeting effi- fine-tuning of enzyme expression. Due to the combinatorial nature of
ciencies (70% compared to approximately 90%) compared to single multigene pathway assembly, a high number of variants is possible,
sgRNA expression (Weninger et al., 2016). thus screening for the best combination is likely becoming the limiting
CRISPR/Cas9-mediated HDR allows the efficient and selection factor. To explore the possible design space, advanced high throughput
marker-free integration of DNA fragments into the genome with the screening methods are needed.
possibility to replace, disrupt or tag a specific genomic locus (Singh For efficient genetic engineering and pathway expression, tunable
et al., 2017). Thereby researches take advantage of the fact that the and differently strong promoters are a prerequisite to drive gene ex-
double-strand break generated by Cas9 recruits the HR machinery and pression. Apart from overexpression of heterologous genes, exchange of
thus increases HR efficiency at the targeted locus. Weninger et al. native promoters with other promoters is a way to overcome feedback
(2018) suggested the use of a ku70 background strain to achieve high inhibition, optimize enzyme abundance and increase the metabolic flux
efficiency of marker-free HDR. However, this reduces transformation towards a product of interest (Liu et al., 2015; Marx et al., 2008). Ef-
efficiencies drastically and is not suited to generate indel mutations. ficient and regulatable P. pastoris promoters are often derived from
The Cas9/sgRNA episomal plasmid described by Gassler et al. (In press) carbon-source utilization pathways such as the methanol utilization
(part of the CRISPi kit available at Addgene #1000000136) and linear pathway (MUT) or the rhamnose-utilization pathway (reviewed by Vogl
donor DNA fragments with flanking regions of approximately 1000 bp and Glieder, 2013). Natively, these promoters are induced by the
each enabled targeting efficiencies above 50% on a routine basis, si- availability of the respective carbon source and shut off on glucose or
milar to indel generation, in a wild type strain. Please note that genes glycerol surplus. Although the AOX1 promoter is still the most com-
essential for growth can still not be deleted by this method, however, in monly used control element for protein production applications, a wide
such a case Cas9-mediated indels (as described above) or dCas9-medi- variety of other MUT promoters have been described and tested re-
ated CRISPR interference (Lian et al., 2018; Qi et al., 2013) might be cently (Gasser et al., 2015; Prielhofer et al., 2017; Vogl et al., 2016).
suitable to diminish gene function. Together with systematically engineered promoter variants (Hartner
For genes especially hard to knock-out such as och1 (Krainer et al., et al., 2008; Yang et al., 2018), and synthetic promoters (Vogl et al.,
2013; Nett and Gerngross, 2003; Vervecken et al., 2004), where also 2014) a large toolbox of synthetic parts is available, which allows
CRISPR/Cas9 only yielded 50% mutation efficiency (Weninger et al., methanol-induced or in some cases also de-repressed gene expression. If
2016), also more time and work intensive indirect methods might be methanol-free expression systems are intended, strong constitutive
considered. Examples are the simultaneous expression of the gene to be promoters such as PGAP or PTEF1 are frequently used (Vogl and Glieder,
knocked out from an episomal plasmid (Chen et al., 2013) or as part of 2013). Alternatively, the PG promoter series which is induced in glu-
a loxP-flanked disruption cassette (Shibui and Hara, 2017), followed by cose-limiting conditions as often encountered during production pro-
curing of it. Another promising approach to increase HR efficiency was cesses (Prielhofer et al., 2013) or the thiamine-regulatable PTHI11 pro-
the addition of hydroxyurea during cell transformation that arrests the moter (Landes et al., 2016) provide for methanol-independent
cells in the S/G2 phase of the cell cycle, where HR is prevalent to NHEJ regulated promoters. Vitamin or amino acid regulated promoters such
(Tsakraklides et al., 2015). as PSER1, PMET3, and PTHR1 also allow for repression and induction of
pathways independent of the carbon source and can be used to speci-
2.3.3. Novel modular cloning strategies and libraries of synthetic parts for fically down-regulate otherwise essential gene functions (Delic et al.,
P. pastoris 2013). Additionally, less strong constitutive or regulatable promoters,
For rapid optimization of single expression cassettes and whole as provided e.g. in the GoldenPiCS toolbox (Prielhofer et al., 2017) can
synthetic pathways, efficient and high throughput modular cloning be of interest for metabolic engineering applications and to balance
strategies are essential. To this end, seamless modular cloning strategies multigene expression in synthetic pathways. Altogether, more than 100
such as Gibson assembly, Golden Gate assembly (GGA), or Restriction promoters have been described for use in P. pastoris.
site free cloning (RSFC) have been adapted and established for use in P. Recently, a non-orthogonal combination of promoter engineering
pastoris. Initially GGA and RSFC were used to design or optimize single (by duplication or deletion of transcription factor binding sites) and

6
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

overexpression of the respective transcription factors (TF) has been quantitative trait loci mapping (Swinnen et al., 2012). The recent de-
reported for PGAP (Ata et al., 2017). A similar approach was followed up velopment of mutant P. pastoris strains with defined stable mating types
for PAOX1, where single overexpression of two out of three different TFs (Heistinger et al., 2018) further enhances the accessibility of the genetic
involved in regulation of the MUT pathway (Mxr1, Mit1) converted P. repertoire of this yeast.
pastoris to methanol-free PAOX1 expression (Vogl et al., 2018b). Con-
currently, it was reported that knockout of three repressing transcrip- 3. Metabolic engineering applications
tion factors (Δmig1Δmig2Δnrg1) in combination with Mit1-over-
expression renders PAOX1 methanol-independent (Wang et al., 2017). As 3.1. Engineering of protein glycosylation
an example of a truly orthogonal system, a β-estradiol-inducible circuit
consisting of an estradiol-inducible zinc-finger transcription factor and Many secretory proteins are glycosylated, and it is no surprise that
corresponding artificial binding sites was shown to be operational in P. glycosylation of recombinant therapeutic proteins is a major quality
pastoris (Perez-Pinera et al., 2016). issue for biopharmaceutical production. After polypeptide translocation
Less effort was so far put into transcription terminators (TT), which into the ER, glycans are either linked to asparagine residues (N-glycans)
have also been implicated with mRNA stability. Recent high- or to serine or threonine residues (O-glycans). While the uniform core
throughput screening in S. cerevisiae emphasized that TTs are important structure of N-glycans consists of two N-acetyl glucosamine and three
regulatory elements which should be considered for the fine-tuning of mannose residues forming a branch, there is a species dependent wide
gene expression and metabolic pathways as terminator selection can variety of sugar structures attached to the core. N-glycans of yeast
influence the flux through a metabolic pathway (Curran et al., 2013; proteins, as of most fungi, mainly consist of mannose residues adjacent
Wei et al., 2017; Yamanishi et al., 2013). Apart from the commonly to the two N-acetyl-glucosamine (GlcNAc) residues, while human pro-
used AOX1-TT and the S. cerevisiae derived CYC1-TT, two recent studies teins contain so-called complex N-glycans consisting of GlcNAc, ga-
provided more insight into this topic in P. pastoris: while Vogl et al. lactose and sialic acid adjacent to the core mannose residues. Concerns
(2016) focused on TTs of MUT genes, Prielhofer et al. (2017) assessed that mannose-rich glycans of yeast made proteins may be immunogenic
the efficiency of TTs of strongly expressed P. pastoris genes including (Jacobs and Callewaert, 2009) as well as the positive impact of complex
many ribosomal protein genes. All tested P. pastoris TTs showed rather glycan pattern on efficacy and stability of therapeutic proteins (Fukuda
similar reporter levels, slightly exceeding ScCYC1-TT by maximum et al., 1989) prompted to engineer N-glycosylation patterns in P. pas-
50%. The so far tested P. pastoris terminators had a size of 174–507 bps toris. In fungi the first committed step towards high-mannose glycan
(Prielhofer et al., 2017; Vogl et al., 2016), whereas significantly shorter formation is an α-1,6 mannosylation by Och1, so that the deletion or
(35–75 bps long) and even synthetic terminator sequences have been inactivation of this enzyme was the first engineering step towards hu-
identified for S. cerevisiae (Curran et al., 2015). As TTs seem to be manization of N-glycans in P. pastoris. After removal of terminal man-
functional across yeast species (Wagner and Alper, 2016), it will be of nose residues by mannosidases, the complex glycan structure is built up
interest to test and implement such short TTs also into the synthetic P. by the consecutive activity of GlcNAc transferases, galactosyl trans-
pastoris toolbox. ferase and sialyl transferase. These steps required complex metabolic
As stated above, the traditional method is to integrate the expres- engineering to provide both the sugar transferases and the precursors
sion cassette(s) into the genome of P. pastoris. Only a few integration (activated sugars) at the right cellular localization, thus ensuring the
loci have been comparatively analyzed so far (3´-AOX1, 5´-AOX1, correct consecutive additions of sugars to the glycans. Targeting of
AOX1, 5´-ENO1, 5´-RGI2, 5´-GAP/TDH3, GUT1, rDNA locus), which all mannosidases and sugar transferases to consecutive locations in the
seemed to allow gene expression from a single expression cassette at secretory pathway was achieved with gene fusion libraries of fungal
similar strength (Prielhofer et al., 2017; Vogl et al., 2018a). membrane protein leader sequences with mannosidases and trans-
A recent approach involving in vivo self-ligation cloning of short ferases of animal and fungal origin, followed by screening for correct
overlapping DNA sequences and episomal vectors (Camattari et al., glycan patterns (Choi et al., 2003; Hamilton et al., 2003, 2006). The
2016) is a promising step towards in vivo single step pathway con- base strain lacking Och1 activity expressed 3 recombinant genes:
struction similar as described for S. cerevisiae (Shao and Zhao, 2009), Kluyveromyces lactis uridine 5′-diphosphate (UDP)-GlcNAc transporter,
however, efficient assembly of larger multi-gene pathways in P. pastoris the mouse mannosidase MnsIA catalytic domain fused to the N-terminal
will need further optimization. Episomal vectors (using e.g. the native localization peptide of the ER protein Sec12 from S. cerevisiae, and
P. pastoris ARS, the K. lactis Pan-ARS or the P. pastoris mitochondrial human GlcNAc transferase GnTI fused to the leader sequence from the
ARS sequences) are also a useful possibility for screening experiments S. cerevisiae Golgi protein Mnn9 (Choi et al., 2003). Additional over-
e.g. for the assessment of activity of enzyme variants, however, they are expression of Drosophila melanogaster mannosidase II (ManII) and rat
not suitable for protein production processes so far, as they require a GlcNAc transferase GnTII, both fused to the N-terminal localization
constant positive selection pressure due to their segregational in- signal of the S. cerevisiae Golgi protein Mnn2 lead to the predominant
stability (Camattari et al., 2016; Liachko and Dunham, 2014; formation of GlcNAc2Man3GlcNAc2 N-glycans (Hamilton et al., 2003).
Schwarzhans et al., 2017b). Addition of galactose was achieved by overexpression of Schizo-
Apart from precise genetic engineering of industrial strains, adap- saccharomyces pombe galactose epimerase, Drosophila melanogaster UDP-
tive laboratory evolution (ALE) is a valuable tool to engineer their Gal transporter, and human β-1,4 galactosyl transferase (Li et al.,
genomes, especially when the desired traits are more complex and ge- 2006). Several further steps needed to be engineered for terminal sia-
netically not well defined. Evolution and subsequent reverse en- lylation (Hamilton et al., 2006). After screening more than 120 per-
gineering proved to be efficient for increasing stress tolerance and mutations of genes involved in sialic acid synthesis, transport and
product levels in many host organisms (Dragosits and Mattanovich, transfer, five enzymes were selected: human UDP-N-acetylglucosamine-
2013). So far, the application of laboratory evolution has been rather 2-epimerase/N-acetylmannosamine kinase (GNE), human N-acet-
limited in P. pastoris, which might be due to the lack of easily selectable ylneuraminate-9-phosphate synthase (SPS), human CMP–sialic acid
traits connected to protein production. In this respect, ALE has been synthase (CSS), mouse CMP–sialic acid transporter (CST), and a library
used to select for enhanced growth on methanol yielding some clones of chimeric sialyltransferases linked to yeast type-II transmembrane
that also produced more heterologous protein (Moser et al., 2017). localization peptides (ST). After some further optimization 90% sialy-
Mating as another approach to engineer genomes without exact lated N-glycans (Sia2Gal2GlcNAc2Man3GlcNAc2) were achieved.
knowledge of the involved genetic traits was not fully accessible for P. Similarly, (Jacobs et al., 2009) described the conversion of a wild
pastoris until recently. Mating type switching and the rapid haploidi- type P. pastoris strain to a strain producing Gal2GlcNAc2Man3GlcNAc2
zation after mating prevented the efficient applications of methods like N-glycans by the consecutive transformation and screening with five so-

7
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

called GlycoSwitch vectors, encoding ManI (Trichoderma reesei man- stress responses (Geiler-Samerotte et al., 2011). Additionally, high
nosidase), GnTI, GalT, ManII, and GnTII, respectively. protein production reduces cell fitness by diverting resources from the
It should be noted that any modification of N-glycosylation also synthesis of macromolecules needed to sustain an optimal growth rate.
affects all native yeast proteins and thus potentially strain fitness. This Depending on the extent of growth rate reduction, this could lead to
aspect was more recently discussed for OCH1 deletion strains (Krainer low productivities, especially if the product is growth-coupled
et al., 2013) which should also affect further engineered strains. (Rebnegger et al., 2014).
However, it was shown that glycoengineered P. pastoris was able to Metabolic engineering strategies have been applied to address the
produce recombinant human erythropoietin (Hamilton et al., 2006) and constraints of protein production described above, by improving the
IgG in the lab and pilot scale (Ye et al., 2011). Recently, a strategy to supply of precursors and increasing the cellular redox and energy effi-
obtain simplified homogeneous glycosylation profiles in recombinant ciency. A study by Nocon et al. (2014) demonstrated the use of genome-
proteins was implemented in P. pastoris. The approach, termed Glyco- scale metabolic modeling to predict gene knock-out and overexpression
delete, is based on the expression of the fungal endoglycosidase endoT, targets in the central carbon metabolism that would increase protein
which specifically cleaves N-glycan structures leaving behind a single production. Many of those in-silico targets were successfully validated
N-GlcNAc attached to the protein (Claes et al., 2016) experimentally in P. pastoris. These included the deletion of genes di-
Yeast O-glycosylation differs from mammalian-type O-glycosylation verting carbon towards fermentative pathways, the overexpression of
as well. In P. pastoris, O-glycans consist of short linear α-1,2-linked malate dehydrogenase, which could increase the supply of mitochon-
mannose chains with potential β-1,2-linked or phosphorylated mannose drial NADH, and the overexpression of the PPP. Further combinatorial
at the outer ends. After deletion of the genes responsible for β-1,2- overexpression of the enzymes in the oxidative part of the PPP resulted
mannosylation and α-1,2-phosphomannosylation, a T. reesei mannosi- in improved titers of protein (Nocon et al., 2016). This could be ex-
dase gene was integrated into P. pastoris, leading to single mannose O- plained by an increased supply of NADPH and precursors (e.g. ery-
glycans (Hamilton et al., 2013). Following the same engineering throse-4-phosphate or ribose-5-phosphate) or a reduced oxidative
strategy for the addition of GlcNAc, galactose and sialic acid as de- stress. Exploring the central role of NADH for protein production in P.
scribed above led to the formation of complex sialylated O-glycans in P. pastoris, a recent study introduced a NADH oxidase from Lactococcus
pastoris (Hamilton et al., 2013). lactis (noxE) to balance the excess of NADH generated from methanol
dissimilation (Jayachandran et al., 2017). The increased thermo-
dynamic efficiency of the pathway yielded a higher methanol uptake, a
3.2. Metabolic engineering to enhance protein production corresponding upregulation of several enzymes in the methanol meta-
bolism, and a 37% higher titer of Lipase B from Candida antarctica
Recombinant protein production is a metabolically costly process. It (CalB). However, NADH consumption by the heterologous oxidase re-
diverts several cellular machineries from their evolutionary goal of cell duced the supply of ATP from oxidative phosphorylation, resulting in a
growth and maintenance, drains precursors from the central carbon lower adenylate energy charge (AEC). Upon expression of the adenylate
metabolism, consumes redox and energy cofactors, and can create en- kinase gene ADK1 from S. cerevisiae the AEC was restored above the
ergetic inefficiencies in the metabolism (Kafri et al., 2016; Klein et al., wild type level, resulting in a 1.5-fold higher titer of secreted CalB
2015; Wu et al., 2016). Given that sufficient gene copies of a protein of protein.
interest are expressed under a strong promoter, the synthesis of high A general metabolic response to high recombinant protein produc-
quality protein requires high cellular capacities for translation, folding, tion is the depletion of intracellular amino acids concentrations
posttranslational modifications and localization of the protein (Fig. 1). (Carnicer et al., 2012b; P. Liu et al., 2016). Thus, the supply of amino
Therefore, any of these molecular machineries and the metabolic re- acids can be engineered to improve protein production in P. pastoris.
sources they consume can become a bottleneck for recombinant protein For example, overexpression of GCN4, encoding a general transcrip-
production. Recycling of stalled cellular machineries and misfolded or tional activator of amino acid biosynthesis, was shown to double the
low quality protein greatly reduces cell energetic efficiency and activate

Fig. 1. Metabolic constraints of recombinant protein pro-


duction. (A) Genome-scale metabolic modeling related to
protein production is yet to include constraints beyond the
metabolic limitations. (B) Recombinant protein produc-
tion drains energy, precursors and redox potential from
the central carbon metabolism across all production steps.

8
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

secretion of a glucose oxidase (Gu et al., 2015) or a fungal carbox-


ylesterase (Gasser et al., 2014). Furthermore, direct overexpression of
metabolic enzymes in the anabolism of serine, isoleucine, alanine and
aromatic amino acids resulted in improvement of 1.2–1.8 fold in pro-
tein accumulation and several fold increases in most of the amino acids
pools (Gasser et al., 2014). Thus, there seems to be a potential to op-
timize the synthesis pathways of amino acids by tuning enzyme abun-
dance or their kinetics (Bar-Even et al., 2011).
The metabolic burden of protein production can be alleviated by
supplying key amino acids, vitamins and fatty acids to the cell meta-
bolism (Heyland et al., 2011; P. Liu et al., 2016; Matthews et al., 2018).
This underlines the tight energetic and redox constraints imposed by
high recombinant protein production. Hence, particularly expensive
macromolecules building the cellular machinery for protein synthesis
and processing could be engineered to free up scarce metabolic re-
sources. For example, modifying the lipid composition of P. pastoris
cells by altering culture conditions has been shown to improve protein
secretion (Adelantado et al., 2017; Baumann et al., 2011). These
changes, which were more pronounced in sphingolipid and sterol
composition, were obtained by applying hypoxic conditions or an in- Fig. 2. Primary and secondary metabolites produced in P. pastoris. Specific
hibitor of ergosterol synthesis. In the future, direct metabolic en- pathways towards products already made in P. pastoris, and to their precursors
gineering of lipid synthesis and degradation pathways could be ex- are highlighted in yellow and orange, respectively. Common metabolic nodes at
plored to fine tune lipid composition for improve protein secretion in P. key precursors (pathways highlighted in red) indicate targets for future en-
pastoris. gineering of chassis strains as platforms for the development of multiple pro-
Besides direct metabolic limitations of protein synthesis, which are duction strains for different metabolic products.
already amenable to metabolic modeling and analysis, folding and se-
cretion have been shown to be potential major bottlenecks of re- constructed by overproduction of three native enzymes diverting pyr-
combinant protein production (Gasser et al., 2008). As in other hosts, uvate into the intermediate 2-ketoisovalerate. Then, the alcohol dehy-
many of these limitations have been addressed by cell engineering drogenase gene ADH7 from S. cerevisiae and a 2-keto acid decarboxylase
(Zahrl et al., 2017), however actual productivities still lag behind those gene from Lactobacillus lactis were expressed to convert 2-ketoisovale-
predicted by metabolic models. Integration of constraints downstream rate into isobutanol. Shake flask cultivations of the engineered strain
of direct metabolic costs of protein synthesis into metabolic models, as accumulated up to 2.2 g/L isobutanol, proving the functionality of the
suggested by Feizi et al. (2013), will therefore be necessary to improve pathway. Furthermore, isobutyl acetate was produced upon expression
the predictions from genome-scale models (Fig. 1), and to enable sys- of the alcohol O-acyltransferase gene ATF1 from S. cerevisiae. Also re-
tems engineering of the synthesis and secretion of proteins. cently, expression of a lactate dehydrogenase gene from Bos taurus
enabled the construction of a strain converting 63% of glycerol into
3.3. Metabolic engineering for the production of metabolites lactic acid, which was secreted by a newly characterized native trans-
porter (de Lima et al., 2016).
Given the proven history of P. pastoris as an industrial cell factory P. pastoris has also a distinct capacity to produce functional cyto-
for protein production, there has been an increasing interest in ex- chrome P450 enzymes (Geier et al., 2012). This has encouraged efforts
ploring its potential to produce primary and secondary metabolites to engineer it as host for the synthesis of plant secondary metabolites. A
(Fig. 2). Many of the published studies harnessed the distinct phy- remarkable example is the production of the valuable fragrance com-
siology and genetic makeup of P. pastoris to introduce heterologous pound (+)-nootkatone from glucose (Wriessnegger et al., 2014). By
enzymes and optimize native metabolic pathways. In 2006, He et al. expressing the membrane associated premnaspirodiene oxygenase gene
(2006) described the overproduction of S-adenosyl-L-methionine from Hyoscyamus muticus and a cytochrome P450 reductase from Ara-
(SAM), probably the first example of metabolic engineering in P. pas- bidopsis thaliana, trans-nootkatol could be efficiently synthesized from
toris. SAM is a component of the sulfur metabolism acting as a methyl supplemented (+)-valencene. Next, an endogenous alcohol dehy-
group donor in several biochemical reactions. It has potential applica- drogenase was identified that efficiently converted trans-nootkatol to
tions in the treatment of many medical conditions including Alzhei- (+)-nootkatone. In order to eliminate the need for (+)-valencene
mer's disease, osteoarthritis and liver disease. To engineer P. pastoris for supplementation, valencene synthase from Callitropsis nootkatensis was
SAM production, conversion of L-methionine to SAM was first improved further introduced, which in combination with a truncated hydroxy-
by overexpression of the native methionine adenosyltransferase gene methylglutaryl-CoA reductase, enabled the direct synthesis of 0.2 g/L
SAM2. Then, the gene encoding the enzyme cystathionine-β-synthase (+)-nootkatone. The potential of P. pastoris to produce pharmaceuti-
was deleted, resulting in higher supply of the precursor homocysteine. cally relevant triterpenoides was also demonstrated with the synthesis
The combined strategies resulted in a 45-fold increase in the production of dammarenediol-II from glucose (Liu et al., 2015). Dammarenediol-II
of SAM and a titer of 4.4 g/L, when methionine was supplied in the is a platform molecule that can be converted into various bioactive
culture. In another study, the natural D-arabitol accumulation of P. ginsenosides by glycosylation and oxidation with cytochrome P450-
pastoris as a redox sink (Baumann et al., 2010) and a protective re- dependent monooxygenases. Simple expression of a dammarenediol-II
sponse to high osmotic conditions (Dragosits et al., 2010) was exploited synthase from Panax ginseng (PaDDS) in P. pastoris resulted in only
to evolve a strain with improved D-arabitol production (Cheng et al., 0.030 mg dammarenediol-II/g dry cell weight (DCW). Hence, in order
2014). The resulting strain was used as platform to express hetero- to boost the supply of the intermediate 2,3-oxidosqualene, ERG1 en-
logous D-arabitol and xylitol dehydrogenases, enabling direct synthesis coding for the rate-limiting enzyme squalene epoxidase was over-
of 0.29 g/L-h of the sweetener xylitol. Similarly, the metabolic pathway expressed, and the competing consumption of 2,3-oxidosqualene for the
of L-valine was harnessed to produce isobutanol and isobutyl acetate synthesis of ergosterol was reduced by downregulation of the native
from glucose (Siripong et al., 2018), in one of the few examples of bulk lanosterol synthase gene ERG7. Finally, the resulting strain reached a
chemical production in P. pastoris. In the study, a strain was first

9
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

25-fold higher accumulation of dammarenediol-II. A subsequent study L of glucaric acid in fed-batch cultures (Y. Liu et al., 2016).
explored the use of a protein scaffold to co-localize the squalene Recently, pathway targeting to the peroxisome has attracted interest
epoxidase and PaDDS, which naturally localizes to subcellular lipid as a metabolic engineering strategy in yeasts, because of the possibility
particles (Zhao et al., 2016). The strategy was shown to double the yield to localize orthogonal metabolic pathways and decrease by-product
obtained in a strain without the scaffold, possibly due to the channeling formation (DeLoache et al., 2016). Methylotrophic yeasts in particular
of 2,3-oxidosqualene towards dammarenediol-II. However, the ob- have evolved peroxisomal compartments to minimize the toxic effects
tained yield was still almost 10-fold lower than in the approach of of formaldehyde and hydrogen peroxide, which may damage proteins
pathway optimization performed by Liu et al. (2015). in the cytosol, and to enhance energy efficiency (van der Klei et al.,
In 2013, Gao and coworkers described the synthesis of 6-methyl- 2006). Upon induction with methanol, peroxisomes dominate the cell
salicylic acid (6MSA), the first polyketide to be produced in P. pastoris volume to contain large amounts of alcohol oxidase and catalase, en-
(Gao et al., 2013). Polyketides form a large family of bioactive and abling methanol assimilation as described earlier (Russmayer et al.,
structurally complex compounds produced as secondary metabolites by 2015a). Another interesting native pathway localized in this compart-
diverse organism including bacteria, fungi and plants. Production of ment is the beta-oxidation of fatty acids, which delivers the key pre-
6MSA in P. pastoris required the functional expression of the 6MSA cursors 3-hydroxyacyl-CoA and acetyl-CoA. Poirier et al. (2002) de-
polyketide synthase gene from Aspergillus terreus as well as the phos- scribed the use of a PHA synthase from Pseudomonas aeruginosa to
phopantetheinyl transferase gene from Aspergillus nidulans (npgA). The convert medium chain length 3-hydroxyacyl-CoA from oleic acid into
latter is needed in yeast to activate the acyl carrier protein (ACP) do- polyhydroxyalkanoates (PHAs), bacterial polyesters that can be used as
main of the heterologous polyketide synthase, enabling the transfer of biodegradable polymers. The expression of the PHA synthase under the
malonyl units to the growing acyl chain. The heterologous pathway, control of the promoter of the native acyl-CoA oxidase gene POX1 en-
consuming cytosolic acetyl-CoA and malonyl-CoA, showed to be func- abled PHA accumulation up to 1% DCW, depending on the substrate
tional in P. pastoris, enabled a titer of 2.2 g/L 6MSA. In a subsequent concentration. Similarly, peroxisomal synthesis of carotenoids, a class
study, the complex mycotoxin citrinin was produced by expressing six of terpenoids used as food colorants, antioxidant and nutraceuticals has
genes from Monascus purpureus under the control of the methanol-in- also been attempted (Bhataya et al., 2009). The authors reported the
duced AOX1 promoter (Xue et al., 2017). Citrinin was detected in a introduction of carotenogenic enzymes from Erwinia uredovora, which
concentration 0.6 mg/L after 24 h of methanol induction, with further correctly localized to the peroxisome (Lee et al., 2009) and resulted in a
cultivation reducing the titer because of the low pH (2.4) in the culture. yield of 4.6 mg/g DCW lycopene. Exploiting a cytosol-localized version
Interestingly, correct identification of an intron in the heterologous of the same pathway, (Araya-Garay et al., 2012b) achieved the synth-
polyketide synthase gene from M. purpureus enabled an efficient release esis of 339 μg/g DCW ß-carotene from glucose, by additional expression
of the ketoaldehyde from the ACP domain, which had previously hin- of gene crtL from Ficus carica. Furthermore, introduction of the β-car-
dered citrinin production (Gao et al., 2013). Indeed, in-silico prediction otene ketolase and β-carotene hydroxylase from Agrobacterium aur-
of introns is still a challenge for the functional expression of natural antiacum enabled the synthesis of astaxanthin (Araya-Garay et al.,
product pathway genes in yeasts (Billingsley et al., 2016). In a recent 2012a), a keto-carotenoid used as a feed supplement in aquaculture.
work, the synthesis of two further polyketides, lovastatin and mon- Besides polyhydroxyalcanoates, other high molecular-weight com-
acolin J, was reported (Liu et al., 2018). Lovastatin and derivative pounds have been produced in P. pastoris. Jeong et al. (2014) reported
compounds of monacolin J are widely used for the treatment of car- the synthesis of hyaluronic acid, a clinically relevant biopolymer by
diovascular diseases because of their cholesterol-lowering activity. For overproducing the native UDP-N-acetylglucosamine pyrophosphorylase
production in P. pastoris, a biosynthetic pathway for dihydromonacolin- and phosphoglucose isomerase, in combination with the heterologous
L (DML) was first constructed by expression of the genes encoding for enzymes hyaluronan synthase 2 and UDP-glucose dehydrogenase. Ex-
lovastatin nonaketide synthase, enoyl reductase and thioesterase from pression optimization of the latter resulted in a strain producing
A. terreus. Further oxidation of DML by heterologous cytochrome P450 0.8–1.7 g/L of large molecular weight hyaluronic acid (2.5 MDa), which
enzymes from A. terreus resulted in monacolin-L and monacolin-J, is required for clinical applications.
which could be converted to lovastatin by heterologous expression of a Considering the ability of P. pastoris to grow on a wide range of
lovastatin diketide synthase gene from the same host. After observing substrates and to secrete functional heterologous enzymes, strategies to
accumulation of intermediate metabolites, the synthesis pathway was degrade and utilize biomass feedstocks are particularly interesting in
split to express the upstream DML synthesis module and the remaining this host. For example, P. pastoris was engineered to grow on cellubiose
pathway in two separate strains to be co-cultured, thereby reducing the and carboxymethyl cellulose by simultaneous production of a fungal
individual metabolic burden. After optimizing the relative strain heterologous B-glucosidase, endo and exoglucanase (Kickenweiz et al.,
abundance, the resulting co-culture produced 250 mg/L of lovastatin 2018). Even though no growth was observed on cellulose, the produced
and 593 mg/L monacolin J in bioreactor cultivations using methanol as enzymes were shown to release glucose at higher temperatures. Fur-
substrate. This novel strategy enabled efficient tuning of the synthesis thermore, construction of a mini-cellulosome by co-localizing a xyla-
pathway by controlling the strain abundance, however it was shown to nase from Clostridium cellulovorans and an endoglucanase from Clos-
be dependent on the diffusion of the key intermediate through the cell tridium thermocellum resulted in a strain producing 1 g/L of ethanol by
membrane. direct biomass hydrolysis (Shin et al., 2015).
Metabolic pathway optimization is generally a very challenging
endeavor because of the complex regulation that cells have evolved to 4. Outlook
maintain homeostasis and robustness (Nielsen and Keasling, 2016).
Direct relief of native regulation mechanisms can thus be used to im- The last decade has seen an impressive increase in molecular and
prove metabolite production in P. pastoris. In a study, the production of cell biology knowledge of P. pastoris, as well as state of the art cell
riboflavin (Vitamin B2) was increased by removing the transcriptional engineering and synthetic biology tools. These novel developments will
regulation of each of the six genes in the riboflavin biosynthesis further support metabolic engineering endeavors to make P. pastoris a
pathway (Marx et al., 2008). The approach required the stepwise re- leading host organisms for metabolite production. However, when
placement of all native promoters with the constitutive GAP promoter, forecasting future applications we should first answer the question why
resulting in a strain accumulating up to 175 mg/L riboflavin in fed- P. pastoris should be used as a metabolic engineering platform at all,
batch cultivation. Similarly, the native post-translational regulation of beside other established hosts such as bacteria or S. cerevisiae, among
the synthesis pathway of glucaric acid was by-passed by expression of a yeasts. To date, the published applications of P. pastoris to produce
myo-inositol oxygenase from mouse, enabling the production of 6.61 g/ metabolites are still limited and rather scattered, including just a subset

10
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

of products demonstrated in other cell factories, whereby mainly proof- Research Promotion Agency FFG (BG, JZ, MGS, DM), and by the
of-concept strains have been reported. Titers obtained were lower than Austrian Federal Ministry for Digital and Economic Affairs (bmwd) and
1 g/L in many cases and thus not close to commercialization stage, the National Foundation for Research, Technology and Development
indicating that this is still an emerging field. However, P. pastoris has (BG).
established in a niche for complex metabolite production. Several fac-
tors could account for this. Firstly, the traditional application of P. References
pastoris as a host for heterologous protein production showed its ability
to produce functional bacterial, fungal, and plant enzymes, a key suc- Adelantado, N., Tarazona, P., Grillitsch, K., García-Ortega, X., Monforte, S., Valero, F.,
cess factor in pathway engineering for high value compounds. Feussner, I., Daum, G., Ferrer, P., 2017. The effect of hypoxia on the lipidome of
recombinant Pichia pastoris. Microb. Cell Fact. 16, 86.
Secondly, the Crabtree-negative phenotype of P. pastoris allows pro- Araya-Garay, J.M., Ageitos, J.M., Vallejo, J.A., Veiga-Crespo, P., Sanchez-Perez, A., Villa,
duction of large amounts of biomass and proteins without significant T.G., 2012a. Construction of a novel Pichia pastoris strain for production of xantho-
fermentation, at the expense of a reduced glucose uptake rate. phylls. AMB Express 2, 24.
Araya-Garay, J.M., Feijoo-Siota, L., Rosa-dos-Santos, F., Veiga-Crespo, P., Villa, T.G.,
Connected to this, higher energy efficiency and a higher flux ratio di- 2012b. Construction of new Pichia pastoris X-33 strains for production of lycopene
verted into the PPP as compared to S. cerevisiae likely provides for more and beta-carotene. Appl. Microbiol. Biotechnol. 93, 2483–2492.
resources to synthesize highly reduced complex metabolites. While part Ata, O., Prielhofer, R., Gasser, B., Mattanovich, D., Calik, P., 2017. Transcriptional en-
gineering of the glyceraldehyde-3-phosphate dehydrogenase promoter for improved
of the disadvantages of Crabtree positive yeasts can be circumvented by
heterologous protein production in Pichia pastoris. Biotechnol. Bioeng. 114,
employing fed-batch cultures this increases costs and complexity of 2319–2327.
large scale production and still cannot cope with the limitations of Aung, H.W., Henry, S.A., Walker, L.P., 2013. Revising the representation of fatty acid,
glycerolipid, and glycerophospholipid metabolism in the consensus model of yeast
energy efficiency in S. cerevisiae and related species. On the contrary,
metabolism. Ind. Biotechnol. (New Rochelle N. Y.) 9, 215–228.
efficient and fast production of primary metabolites directly derived Bar-Even, A., Noor, E., Savir, Y., Liebermeister, W., Davidi, D., Tawfik, D.S., Milo, R.,
from the central carbon metabolism is potentially less favored in P. 2011. The moderately efficient enzyme: evolutionary and physicochemical trends
pastoris than in Crabtree positive yeasts, as the central metabolism is shaping enzyme parameters. Biochemistry 50, 4402–4410.
Baumann, K., Carnicer, M., Dragosits, M., Graf, A.B., Stadlmann, J., Jouhten, P.,
tightly controlled and limited. Maaheimo, H., Gasser, B., Albiol, J., Mattanovich, D., Ferrer, P., 2010. A multi-level
Synthetic biology calls for the development of chassis strains as study of recombinant Pichia pastoris in different oxygen conditions. BMC Syst. Biol. 4,
starting platforms for the development of individual production strains. 141.
Baumann, K., Adelantado, N., Lang, C., Mattanovich, D., Ferrer, P., 2011. Protein traf-
Specifically, the synthesis of secondary metabolites initiates from me- ficking, ergosterol biosynthesis and membrane physics impact recombinant protein
tabolic nodes at key intermediate metabolites. Strains dedicated to secretion in Pichia pastoris. Microb. Cell Fact. 10, 93.
overproduce these intermediates constitute such chassis strains. By this Berkhout, J., Bosdriesz, E., Nikerel, E., Molenaar, D., de Ridder, D., Teusink, B.,
Bruggeman, F.J., 2013. How biochemical constraints of cellular growth shape evo-
concept repetitive redesign and optimization of the same pathways can lutionary adaptations in metabolism. Genetics 194, 505–512.
be avoided, as demonstrated in S. cerevisiae (Jouhten et al., 2016). As a Bhataya, A., Schmidt-Dannert, C., Lee, P.C., 2009. Metabolic engineering of Pichia pastoris
special feature of P. pastoris, the highly expandable peroxisomal com- X-33 for lycopene production. Process Biochem. 44, 1095–1102.
Billingsley, J.M., DeNicola, A.B., Tang, Y., 2016. Technology development for natural
partment could be interesting to localize entire heterologous pathways.
product biosynthesis in Saccharomyces cerevisiae. Curr. Opin. Biotechnol. 42, 74–83.
A possible advantage of this strategy would be increased local meta- Camattari, A., Goh, A., Yip, L.Y., Tan, A.H., Ng, S.W., Tran, A., Liu, G., Liachko, I.,
bolite concentrations, which improve the thermodynamic efficiency of Dunham, M.J., Rancati, G., 2016. Characterization of a panARS-based episomal
vector in the methylotrophic yeast Pichia pastoris for recombinant protein production
pathways. Furthermore, energy production by methanol dissimilation
and synthetic biology applications. Microb. Cell Fact. 15, 139.
offers an opportunity to decouple product formation from growth, and Carnicer, M., Baumann, K., Töplitz, I., Sánchez-Ferrando, F., Mattanovich, D., Ferrer, P.,
even from energy metabolism as such, possibly implementing com- Albiol, J., 2009. Macromolecular and elemental composition analysis and extra-
pletely orthogonal anabolic pathways (Pandit et al., 2017). cellular metabolite balances of Pichia pastoris growing at different oxygen levels.
Microb. Cell Fact. 8, 65.
Besides the development of specialized chassis for high value me- Carnicer, M., Canelas, A.B., Ten Pierick, A., Zeng, Z., van Dam, J., Albiol, J., Ferrer, P.,
tabolites, decades of experience with heterologous protein production Heijnen, J.J., van Gulik, W., 2012a. Development of quantitative metabolomics for
in P. pastoris enabled the recent successes of metabolic and cell en- Pichia pastoris. Metabolomics 8, 284–298.
Carnicer, M., Ten Pierick, A., van Dam, J., Heijnen, J.J., Albiol, J., van Gulik, W., Ferrer,
gineering to boost protein productivity. Novel developments of P. pas- P., 2012b. Quantitative metabolomics analysis of amino acid metabolism in re-
toris systems and synthetic biology allow to anticipate a new generation combinant Pichia pastoris under different oxygen availability conditions. Microb. Cell
of dedicated “protein factory” strains with high productivity, also en- Fact. 11, 83.
Caspeta, L., Shoaie, S., Agren, R., Nookaew, I., Nielsen, J., 2012. Genome-scale metabolic
abling the production of hitherto “difficult to express” proteins which reconstructions of Pichia stipitis and Pichia pastoris and in silico evaluation of their
regularly abscond from entering application tests and clinical trials due potentials. BMC Syst. Biol. 6, 24.
to the inability to produce them in reasonable amounts. Chen, Z., Sun, H., Li, P., He, N., Zhu, T., Li, Y., 2013. Enhancement of the gene targeting
efficiency of non-conventional yeasts by increasing genetic redundancy. PLoS One 8,
We anticipate that recent developments in synthetic biology, me- e57952.
tabolic modeling and analysis will accelerate attaining these goals. Cheng, H., Lv, J., Wang, H., Wang, B., Li, Z., Deng, Z., 2014. Genetically engineered Pichia
Several tools for genome engineering, modular part assembly, and pastoris yeast for conversion of glucose to xylitol by a single-fermentation process.
Appl. Microbiol. Biotechnol. 98, 3539–3552.
stable integration will have a great impact on the speed of strain en-
Choi, B.K., Bobrowicz, P., Davidson, R.C., Hamilton, S.R., Kung, D.H., Li, H., Miele, R.G.,
gineering. Furthermore, as metabolic models improve to include further Nett, J.H., Wildt, S., Gerngross, T.U., 2003. Use of combinatorial genetic libraries to
constraints including resource allocation and post-translational pro- humanize N-linked glycosylation in the yeast Pichia pastoris. Proc. Natl. Acad. Sci.
cesses, rational cell refactoring including new targets in regulatory and USA 100, 5022–5027.
Chung, B.K., Selvarasu, S., Andrea, C., Ryu, J., Lee, H., Ahn, J., Lee, D.Y., 2010. Genome-
metabolic genes will become feasible. scale metabolic reconstruction and in silico analysis of methylotrophic yeast Pichia
pastoris for strain improvement. Microb. Cell Fact. 9, 50.
Acknowledgements Claes, K., Vandewalle, K., Laukens, B., Laeremans, T., Vosters, O., Langer, I., Parmentier,
M., Steyaert, J., Callewaert, N., 2016. Modular integrated secretory system en-
gineering in Pichia pastoris to enhance G-protein coupled receptor expression. ACS
DAP was funded by the Austrian Science Fund (FWF): Doctoral Synth. Biol. 5, 1070–1075.
Program BioToP—Biomolecular Technology of Proteins (FWF W1224). Cregg, J.M., Madden, K.R., 1989. Use of site-specific recombination to regenerate se-
lectable markers. Mol. Gen. Genet. 219, 320–323.
Research on P. pastoris in our labs is supported by the Federal Ministry Cregg, J.M., Russell, K.A., 1998. Transformation. Methods Mol. Biol. 103, 27–39.
for Digital and Economic Affairs (bmwd), the Federal Ministry for Cregg, J.M., Barringer, K.J., Hessler, A.Y., Madden, K.R., 1985. Pichia pastoris as a host
Transport, Innovation and Technology (bmvit), the Styrian Business system for transformations. Mol. Cell Biol. 5, 3376–3385.
Cregg, J.M., Madden, K.R., Barringer, K.J., Thill, G.P., Stillman, C.A., 1989. Functional
Promotion Agency SFG, the Standortagentur Tirol, the Government of characterization of the two alcohol oxidase genes from the yeast Pichia pastoris. Mol.
Lower Austria and ZIT – Technology Agency of the City of Vienna Cell Biol. 9, 1316–1323.
through the COMET-Funding Program managed by the Austrian Curran, K.A., Karim, A.S., Gupta, A., Alper, H.S., 2013. Use of expression-enhancing

11
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

terminators in Saccharomyces cerevisiae to increase mRNA half-life and improve gene Hagman, A., Säll, T., Piškur, J., 2014. Analysis of the yeast short-term Crabtree effect and
expression control for metabolic engineering applications. Metab. Eng. 19, 88–97. its origin. FEBS J. 281, 4805–4814.
Curran, K.A., Morse, N.J., Markham, K.A., Wagman, A.M., Gupta, A., Alper, H.S., 2015. Hamilton, S.R., Bobrowicz, P., Bobrowicz, B., Davidson, R.C., Li, H., Mitchell, T., Nett,
Short synthetic terminators for improved heterologous gene expression in yeast. ACS J.H., Rausch, S., Stadheim, T.A., Wischnewski, H., Wildt, S., Gerngross, T.U., 2003.
Synth. Biol. 4, 824–832. Production of complex human glycoproteins in yeast. Science 301, 1244–1246.
Da Silva, N.A., Srikrishnan, S., 2012. Introduction and expression of genes for metabolic Hamilton, S.R., Davidson, R.C., Sethuraman, N., Nett, J.H., Jiang, Y., Rios, S., Bobrowicz,
engineering applications in Saccharomyces cerevisiae. FEMS Yeast Res. 12, 197–214. P., Stadheim, T.A., Li, H., Choi, B.K., Hopkins, D., Wischnewski, H., Roser, J.,
de Lima, P.B., Mulder, K.C., Melo, N.T., Carvalho, L.S., Menino, G.S., Mulinari, E., de Mitchell, T., Strawbridge, R.R., Hoopes, J., Wildt, S., Gerngross, T.U., 2006.
Castro, V.H., Dos Reis, T.F., Goldman, G.H., Magalhães, B.S., Parachin, N.S., 2016. Humanization of yeast to produce complex terminally sialylated glycoproteins.
Novel homologous lactate transporter improves L-lactic acid production from gly- Science 313, 1441–1443.
cerol in recombinant strains of Pichia pastoris. Microb. Cell Fact. 15, 158. Hamilton, S.R., Cook, W.J., Gomathinayagam, S., Burnina, I., Bukowski, J., Hopkins, D.,
De Schutter, K., Lin, Y.C., Tiels, P., Van Hecke, A., Glinka, S., Weber-Lehmann, J., Rouzé, Schwartz, S., Du, M., Sharkey, N.J., Bobrowicz, P., Wildt, S., Li, H., Stadheim, T.A.,
P., Van de Peer, Y., Callewaert, N., 2009. Genome sequence of the recombinant Nett, J.H., 2013. Production of sialylated O-linked glycans in Pichia pastoris.
protein production host Pichia pastoris. Nat. Biotechnol. 27, 561–566. Glycobiology 23, 1192–1203.
Delic, M., Mattanovich, D., Gasser, B., 2013. Repressible promoters – a novel tool to Hartner, F.S., Ruth, C., Langenegger, D., Johnson, S.N., Hyka, P., Lin-Cereghino, G.P., Lin-
generate conditional mutants in Pichia pastoris. Microb. Cell Fact. 12, 6. Cereghino, J., Kovar, K., Cregg, J.M., Glieder, A., 2008. Promoter library designed for
DeLoache, W.C., Russ, Z.N., Dueber, J.E., 2016. Towards repurposing the yeast peroxi- fine-tuned gene expression in Pichia pastoris. Nucleic Acids Res. 36, e76.
some for compartmentalizing heterologous metabolic pathways. Nat. Commun. 7, He, J., Deng, J., Zheng, Y., Gu, J., 2006. A synergistic effect on the production of S-
11152. adenosyl-L-methionine in Pichia pastoris by knocking in of S-adenosyl-L-methionine
DiCarlo, J.E., Norville, J.E., Mali, P., Rios, X., Aach, J., Church, G.M., 2013. Genome synthase and knocking out of cystathionine-beta synthase. J. Biotechnol. 126,
engineering in Saccharomyces cerevisiae using CRISPR-Cas systems. Nucleic Acids Res. 519–527.
41, 4336–4343. Heiss, S., Maurer, M., Hahn, R., Mattanovich, D., Gasser, B., 2013. Identification and
Dragosits, M., Mattanovich, D., 2013. Adaptive laboratory evolution – principles and deletion of the major secreted protein of Pichia pastoris. Appl. Microbiol. Biotechnol.
applications for biotechnology. Microb. Cell Fact. 12, 64. 97, 1241–1249.
Dragosits, M., Stadlmann, J., Graf, A., Gasser, B., Maurer, M., Sauer, M., Kreil, D., Heistinger, L., Gasser, B., Mattanovich, D., 2018. Creation of stable heterothallic strains of
Altmann, F., Mattanovich, D., 2010. The response to unfolded protein is involved in Komagataella phaffii enables dissection of mating gene regulation. Mol. Cell Biol. 38
osmotolerance of Pichia pastoris. BMC Genom. 11, 207. (e00398-17).
Feizi, A., Österlund, T., Petranovic, D., Bordel, S., Nielsen, J., 2013. Genome-scale Heyland, J., Fu, J., Blank, L.M., Schmid, A., 2010. Quantitative physiology of Pichia
modeling of the protein secretory machinery in yeast. PLoS One 8, e63284. pastoris during glucose-limited high-cell density fed-batch cultivation for re-
Fiehn, O., 2002. Metabolomics–the link between genotypes and phenotypes. Plant Mol. combinant protein production. Biotechnol. Bioeng. 107, 357–368.
Biol. 48, 155–171. Heyland, J., Fu, J., Blank, L.M., Schmid, A., 2011. Carbon metabolism limits recombinant
Förster, J., Halbfeld, C., Zimmermann, M., Blank, L.M., 2014. A blueprint of the amino protein production in Pichia pastoris. Biotechnol. Bioeng. 108, 1942–1953.
acid biosynthesis network of hemiascomycetes. FEMS Yeast Res. 14, 1090–1100. Höffner, K., Harwood, S.M., Barton, P.I., 2013. A reliable simulator for dynamic flux
Fukuda, M.N., Sasaki, H., Lopez, L., Fukuda, M., 1989. Survival of recombinant ery- balance analysis. Biotechnol. Bioeng. 110, 792–802.
thropoietin in the circulation: the role of carbohydrates. Blood 73, 84–89. Hong, I.P., Anderson, S., Choi, S.G., 2006. Evaluation of a new episomal vector based on
Gao, L., Cai, M., Shen, W., Xiao, S., Zhou, X., Zhang, Y., 2013. Engineered fungal poly- the GAP promoter for structural genomics in Pichia pastoris. J. Microbiol. Biotechnol.
ketide biosynthesis in Pichia pastoris: a potential excellent host for polyketide pro- 16, 1362–1368.
duction. Microb. Cell Fact. 12, 77. Irani, Z.A., Kerkhoven, E.J., Shojaosadati, S.A., Nielsen, J., 2016. Genome-scale metabolic
Gasser, B., Saloheimo, M., Rinas, U., Dragosits, M., Rodríguez-Carmona, E., Baumann, K., model of Pichia pastoris with native and humanized glycosylation of recombinant
Giuliani, M., Parrilli, E., Branduardi, P., Lang, C., Porro, D., Ferrer, P., Tutino, M., proteins. Biotechnol. Bioeng. 113, 961–969.
Mattanovich, D., Villaverde, A., 2008. Protein folding and conformational stress in Jacobs, P.P., Callewaert, N., 2009. N-glycosylation engineering of biopharmaceutical
microbial cells producing recombinant proteins: a host comparative overview. expression systems. Curr. Mol. Med. 9, 774–800.
Microb. Cell Fact. 7, 11. Jacobs, P.P., Geysens, S., Vervecken, W., Contreras, R., Callewaert, N., 2009. Engineering
Gasser, B., Prielhofer, R., Marx, H., Maurer, M., Nocon, J., Steiger, M., Puxbaum, V., complex-type N-glycosylation in Pichia pastoris using GlycoSwitch technology. Nat.
Sauer, M., Mattanovich, D., 2013. Pichia pastoris: protein production host and model Protoc. 4, 58–70.
organism for biomedical research. Fut. Microbiol. 8, 191–208. Jayachandran, C., Palanisamy Athiyaman, B., Sankaranarayanan, M., 2017. Cofactor
Gasser, B., Steiger, M.G., Mattanovich, D., 2015. Methanol regulated yeast promoters: engineering improved CALB production in Pichia pastoris through heterologous ex-
production vehicles and toolbox for synthetic biology. Microb. Cell Fact. 14, 196. pression of NADH oxidase and adenylate kinase. PLoS One 12, e0181370.
Gasser, B., Mattanovich, D., Sauer, M., Russmayer, H., 2014. Improved Protein Jeong, E., Shim, W.Y., Kim, J.H., 2014. Metabolic engineering of Pichia pastoris for pro-
Production. European Patent EP2952584A1. duction of hyaluronic acid with high molecular weight. J. Biotechnol. 185, 28–36.
Gassler, T., Heistinger, L., Mattanovich, D., Gasser, B., Prielhofer, R., 2018. CRISPR/Cas9- Jorda, J., Suarez, C., Carnicer, M., ten Pierick, A., Heijnen, J.J., van Gulik, W., Ferrer, P.,
mediated homology directed genome editing in Pichia pastoris. Methods Mol. Biol (In Albiol, J., Wahl, A., 2013. Glucose-methanol co-utilization in Pichia pastoris studied
press). by metabolomics and instationary 13C flux analysis. BMC Syst. Biol. 7, 17.
Geier, M., Braun, A., Emmerstorfer, A., Pichler, H., Glieder, A., 2012. Production of Jorda, J., Rojas, H.C., Carnicer, M., Wahl, A., Ferrer, P., Albiol, J., 2014. Quantitative
human cytochrome P450 2D6 drug metabolites with recombinant microbes–a com- metabolomics and instationary 13C-metabolic flux analysis reveals impact of re-
parative study. Biotechnol. J. 7, 1346–1358. combinant protein production on trehalose and energy metabolism in Pichia pastoris.
Geier, M., Fauland, P., Vogl, T., Glieder, A., 2015. Compact multi-enzyme pathways in P. Metabolites 4, 281–299.
pastoris. Chem. Commun. (Camb.). 51, 1643–1646. Jordà, J., Jouhten, P., Cámara, E., Maaheimo, H., Albiol, J., Ferrer, P., 2012. Metabolic
Geiler-Samerotte, K.A., Dion, M.F., Budnik, B.A., Wang, S.M., Hartl, D.L., Drummond, flux profiling of recombinant protein secreting Pichia pastoris growing on glucose:
D.A., 2011. Misfolded proteins impose a dosage-dependent fitness cost and trigger a methanol mixtures. Microb. Cell Fact. 11, 57.
cytosolic unfolded protein response in yeast. Proc. Natl. Acad. Sci. USA 108, Jouhten, P., Boruta, T., Andrejev, S., Pereira, F., Rocha, I., Patil, K.R., 2016. Yeast me-
680–685. tabolic chassis designs for diverse biotechnological products. Sci. Rep. 6, 29694.
Gleeson, M.A., White, C.E., Meininger, D.P., Komives, E.A., 1998. Generation of protease- Kafri, M., Metzl-Raz, E., Jona, G., Barkai, N., 2016. The cost of protein production. Cell
deficient strains and their use in heterologous protein expression. Methods Mol. Biol. Rep. 14, 22–31.
103, 81–94. Kickenweiz, T., Glieder, A., Wu, J.C., 2018. Construction of a cellulose-metabolizing
Goelzer, A., Fromion, V., 2011. Bacterial growth rate reflects a bottleneck in resource Komagataella phaffii (Pichia pastoris) by co-expressing glucanases and β-glucosidase.
allocation. Biochim. Biophys. Acta 1810, 978–988. Appl. Microbiol. Biotechnol. 102, 1297–1306.
Goelzer, A., Muntel, J., Chubukov, V., Jules, M., Prestel, E., Nölker, R., Mariadassou, M., Klein, T., Niklas, J., Heinzle, E., 2015. Engineering the supply chain for protein produc-
Aymerich, S., Hecker, M., Noirot, P., Becher, D., Fromion, V., 2015. Quantitative tion/secretion in yeasts and mammalian cells. J. Ind. Microbiol. Biotechnol. 42,
prediction of genome-wide resource allocation in bacteria. Metab. Eng. 32, 232–243. 453–464.
Goffeau, A., Barrell, B.G., Bussey, H., Davis, R.W., Dujon, B., Feldmann, H., Galibert, F., Krainer, F.W., Gmeiner, C., Neutsch, L., Windwarder, M., Pletzenauer, R., Herwig, C.,
Hoheisel, J.D., Jacq, C., Johnston, M., Louis, E.J., Mewes, H.W., Murakami, Y., Altmann, F., Glieder, A., Spadiut, O., 2013. Knockout of an endogenous mannosyl-
Philippsen, P., Tettelin, H., Oliver, S.G., 1996. Life with 6000 genes. Science 274 transferase increases the homogeneity of glycoproteins produced in Pichia pastoris.
(546), 563–567. Sci. Rep. 3, 3279.
Gombert, A.K., Moreira dos Santos, M., Christensen, B., Nielsen, J., 2001. Network Kuberl, A., Schneider, J., Thallinger, G.G., Anderl, I., Wibberg, D., Hajek, T., Jaenicke, S.,
identification and flux quantification in the central metabolism of Saccharomyces Brinkrolf, K., Goesmann, A., Szczepanowski, R., Puhler, A., Schwab, H., Glieder, A.,
cerevisiae under different conditions of glucose repression. J. Bacteriol. 183, Pichler, H., 2011. High-quality genome sequence of Pichia pastoris CBS7435. J.
1441–1451. Biotechnol. 154, 312–320.
Gu, L., Zhang, J., Liu, B., Du, G., Chen, J., 2015. High-level extracellular production of Kurtzman, C.P., 2005. Description of Komagataella phaffii sp. nov. and the transfer of
glucose oxidase by recombinant Pichia pastoris using a combined strategy. Appl. Pichia pseudopastoris to the methylotrophic yeast genus Komagataella. Int. J. Syst.
Biochem. Biotechnol. 175, 1429–1447. Evol. Microbiol. 55, 973–976.
Guilliermond, A., 1920. Zygosaccharomyces pastori, nouvelle espèce de levures copulation Landes, N., Gasser, B., Vorauer-Uhl, K., Lhota, G., Mattanovich, D., Maurer, M., 2016. The
hétérogamique. Bull. Soc. Mycol. Fr. 36, 203–211. vitamin-sensitive promoter PTHI11 enables pre-defined autonomous induction of re-
Hackett, S.R., Zanotelli, V.R., Xu, W., Goya, J., Park, J.O., Perlman, D.H., Gibney, P.A., combinant protein production in Pichia pastoris. Biotechnol. Bioeng. 113, 2633–2643.
Botstein, D., Storey, J.D., Rabinowitz, J.D., 2016. Systems-level analysis of me- Laukens, B., De Visscher, C., Callewaert, N., 2015. Engineering yeast for producing
chanisms regulating yeast metabolic flux. Science 354, aaf2786. human glycoproteins: where are we now? Future Microbiol. 10, 21–34.

12
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

Lee, P.C., Yoon, Y., Schmidt-Dannert, C., 2009. Investigation of cellular targeting of manipulations. Biotechnol. Bioeng. 58, 125–132.
carotenoid pathway enzymes in Pichia pastoris. J. Biotechnol. 140, 227–233. Nielsen, J., Keasling, J.D., 2016. Engineering cellular metabolism. Cell 164, 1185–1197.
Lehnen, M., Ebert, B.E., Blank, L.M., 2017. A comprehensive evaluation of constraining Nocon, J., Steiger, M.G., Pfeffer, M., Sohn, S.B., Kim, T.Y., Maurer, M., Russmayer, H.,
amino acid biosynthesis in compartmented models for metabolic flux analysis. Metab. Pflugl, S., Ask, M., Haberhauer-Troyer, C., Ortmayr, K., Hann, S., Koellensperger, G.,
Eng. Commun. 5, 34–44. Gasser, B., Lee, S.Y., Mattanovich, D., 2014. Model based engineering of Pichia pas-
Lewis, N.E., Nagarajan, H., Palsson, B.O., 2012. Constraining the metabolic genotype- toris central metabolism enhances recombinant protein production. Metab. Eng. 24,
phenotype relationship using a phylogeny of in silico methods. Nat. Rev. Microbiol. 129–138.
10, 291–305. Nocon, J., Steiger, M., Mairinger, T., Hohlweg, J., Russmayer, H., Hann, S., Gasser, B.,
Li, H., Sethuraman, N., Stadheim, T., Zha, D., Prinz, B., Ballew, N., Bobrowicz, P., Choi, Mattanovich, D., 2016. Increasing pentose phosphate pathway flux enhances re-
B., Cook, W., Cukan, M., Houston-Cummings, N., Davidson, R., Gong, B., Hamilton, combinant protein production in Pichia pastoris. Appl. Microbiol. Biotechnol. 100,
S., Hoopes, J., Jiang, Y., Kim, N., Mansfield, R., Nett, J., Rios, S., Strawbridge, R., 5955–5963.
Wildt, S., Gerngross, T., 2006. Optimization of humanized IgGs in glycoengineered O'Brien, E.J., Palsson, B.O., 2015. Computing the functional proteome: recent progress
Pichia pastoris. Nat. Biotechnol. 24, 210–215. and future prospects for genome-scale models. Curr. Opin. Biotechnol. 34, 125–134.
Liachko, I., Dunham, M.J., 2014. An autonomously replicating sequence for use in a wide O'Brien, E.J., Lerman, J.A., Chang, R.L., Hyduke, D.R., Palsson, B., 2013. Genome-scale
range of budding yeasts. FEMS Yeast Res. 14, 364–367. models of metabolism and gene expression extend and refine growth phenotype
Lian, J., HamediRad, M., Zhao, H., 2018. Advancing metabolic engineering of prediction. Mol. Syst. Biol. 9, 693.
Saccharomyces cerevisiae using the CRISPR/Cas System. Biotechnol. J. http://dx.doi. Obst, U., Lu, T.K., Sieber, V., 2017. A modular toolkit for generating Pichia pastoris se-
org/10.1002/biot.201700601. cretion libraries. ACS Synth. Biol. 6, 1016–1025.
Liu, P., Huang, M., Guo, M., Qian, J., Lin, W., Chu, J., Zhuang, Y., Zhang, S., 2016. Pan, R., Zhang, J., Shen, W.L., Tao, Z.Q., Li, S.P., Yan, X., 2011. Sequential deletion of
Combined 13C-assisted metabolomics and metabolic flux analysis reveals the impacts Pichia pastoris genes by a self-excisable cassette. FEMS Yeast Res. 11, 292–298.
of glutamate on the central metabolism of high beta-galactosidase-producing Pichia Pandit, A.V., Srinivasan, S., Mahadevan, R., 2017. Redesigning metabolism based on
pastoris. Bioresour. Bioprocess. 3, 47. orthogonality principles. Nat. Commun. 8, 15188.
Liu, X.B., Liu, M., Tao, X.Y., Zhang, Z.X., Wang, F.Q., Wei, D.Z., 2015. Metabolic en- Perez-Pinera, P., Han, N., Cleto, S., Cao, J., Purcell, O., Shah, K.A., Lee, K., Ram, R., Lu,
gineering of Pichia pastoris for the production of dammarenediol-II. J. Biotechnol. T.K., 2016. Synthetic biology and microbioreactor platforms for programmable
216, 47–55. production of biologics at the point-of-care. Nat. Commun. 7, 12211.
Liu, Y., Gong, X., Wang, C., Du, G., Chen, J., Kang, Z., 2016. Production of glucaric acid Phaff, H., Miller, M., Shifrine, M., 1956. The taxonomy of yeasts isolated from Drosophila
from myo-inositol in engineered Pichia pastoris. Enzym. Microb. Technol. 91, 8–16. in the Yosemite region of California. Antonie Van Leeuwenhoek 22, 145–161.
Liu, Y., Tu, X., Xu, Q., Bai, C., Kong, C., Liu, Q., Yu, J., Peng, Q., Zhou, X., Zhang, Y., Cai, Piva, L.C., Bentacur, M.O., Reis, V.C.B., De Marco, J.L., Moraes, L.M.P., Torres, F.A.G.,
M., 2018. Engineered monoculture and co-culture of methylotrophic yeast for de novo 2017. Molecular strategies to increase the levels of heterologous transcripts in
production of monacolin J and lovastatin from methanol. Metab. Eng. 45, 189–199. Komagataella phaffii for protein production. Bioengineered 8, 441–445.
Love, K.R., Shah, K.A., Whittaker, C.A., Wu, J., Bartlett, M.C., Ma, D., Leeson, R.L., Priest, Poirier, Y., Erard, N., MacDonald-Comber Petetot, J., 2002. Synthesis of poly-
M., Borowsky, J., Young, S.K., Love, J.C., 2016. Comparative genomics and tran- hydroxyalkanoate in the peroxisome of Pichia pastoris. FEMS Microbiol Lett. 207,
scriptomics of Pichia pastoris. BMC Genom. 17, 550. 97–102.
Lu, W., Su, X., Klein, M.S., Lewis, I.A., Fiehn, O., Rabinowitz, J.D., 2017. Metabolite Prielhofer, R., Maurer, M., Klein, J., Wenger, J., Kiziak, C., Gasser, B., Mattanovich, D.,
measurement: pitfalls to avoid and practices to follow. Annu. Rev. Biochem. 86, 2013. Induction without methanol: novel regulated promoters enable high-level ex-
277–304. pression in Pichia pastoris. Microb. Cell Fact. 12, 5.
Maaheimo, H., Fiaux, J., Cakar, Z.P., Bailey, J.E., Sauer, U., Szyperski, T., 2001. Central Prielhofer, R., Barrero, J.J., Steuer, S., Gassler, T., Zahrl, R., Baumann, K., Sauer, M.,
carbon metabolism of Saccharomyces cerevisiae explored by biosynthetic fractional Mattanovich, D., Gasser, B., Marx, H., 2017. GoldenPiCS: a golden gate-derived
13
C labeling of common amino acids. Eur. J. Biochem. 268, 2464–2479. modular cloning system for applied synthetic biology in the yeast Pichia pastoris. BMC
Machado, D., Herrgård, M., 2014. Systematic evaluation of methods for integration of Syst. Biol. 11, 123.
transcriptomic data into constraint-based models of metabolism. PLoS Comput. Biol. Qi, L.S., Larson, M.H., Gilbert, L.A., Doudna, J.A., Weissman, J.S., Arkin, A.P., Lim, W.A.,
10, e1003580. 2013. Repurposing CRISPR as an RNA-guided platform for sequence-specific control
Mairinger, T., Steiger, M., Nocon, J., Mattanovich, D., Koellensperger, G., Hann, S., 2015. of gene expression. Cell 152, 1173–1183.
Gas chromatography-quadrupole time-of-flight mass spectrometry-based determina- Raschmanová, H., Weninger, A., Glieder, A., Kovar, K., Vogl, T., 2018. Implementing
tion of isotopologue and tandem mass isotopomer fractions of primary metabolites CRISPR-Cas technologies in conventional and non-conventional yeasts: current state
for 13C-metabolic flux analysis. Anal. Chem. 87, 11792–11802. and future prospects. Biotechnol. Adv. http://dx.doi.org/10.1016/j.biotechadv.2018.
Mairinger, T., Wegscheider, W., Peña, D.A., Steiger, M.G., Koellensperger, G., Zanghellini, 01.006.
J., Hann, S., 2018. Comprehensive assessment of measurement uncertainty in 13C- Rebnegger, C., Graf, A.B., Valli, M., Steiger, M.G., Gasser, B., Maurer, M., Mattanovich,
based metabolic flux experiments. Anal. Bioanal. Chem. http://dx.doi.org/10.1007/ D., 2014. Pichia pastoris, growth rate regulates protein synthesis and secretion,
s00216-018-1017-7. mating and stress response. Biotechnol. J. 9, 511–525.
Marx, H., Mattanovich, D., Sauer, M., 2008. Overexpression of the riboflavin biosynthetic Rebnegger, C., Vos, T., Graf, A.B., Valli, M., Pronk, J.T., Daran-Lapujade, P., Mattanovich,
pathway in Pichia pastoris. Microb. Cell Fact. 7, 23. D., 2016. Pichia pastoris exhibits high viability and a low maintenance energy re-
Mattanovich, D., Graf, A., Stadlmann, J., Dragosits, M., Redl, A., Maurer, M., Kleinheinz, quirement at near-zero specific growth rates. Appl. Environ. Microbiol. 82,
M., Sauer, M., Altmann, F., Gasser, B., 2009. Genome, secretome and glucose trans- 4570–4583.
port highlight unique features of the protein production host Pichia pastoris. Microb. Russmayer, H., Buchetics, M., Gruber, C., Valli, M., Grillitsch, K., Modarres, G., Guerrasio,
Cell Fact. 8, 29. R., Klavins, K., Neubauer, S., Drexler, H., Steiger, M., Troyer, C., Al Chalabi, A.,
Mattanovich, M., Russmayer, H., Scharl-Hirsch, T., Puxbaum, V., Burgard, J., Krebiehl, G., Sonntag, D., Zellnig, G., Daum, G., Graf, A.B., Altmann, F.,
Mattanovich, D., Hann, S., 2017. Metabolomics of Pichia pastoris: impact of buffering Koellensperger, G., Hann, S., Sauer, M., Mattanovich, D., Gasser, B., 2015a. Systems-
conditions on the kinetics and nature of metabolite loss during quenching. FEMS level organization of yeast methylotrophic lifestyle. BMC Biol. 13, 80.
Yeast Res. 17, fox016. Russmayer, H., Troyer, C., Neubauer, S., Steiger, M.G., Gasser, B., Hann, S.,
Matthews, C.B., Kuo, A., Love, K.R., Love, J.C., 2018. Development of a general defined Koellensperger, G., Sauer, M., Mattanovich, D., 2015b. Metabolomics sampling of
medium for Pichia pastoris. Biotechnol. Bioeng. 115, 103–113. Pichia pastoris revisited: rapid filtration prevents metabolite loss during quenching.
Molenaar, D., van Berlo, R., de Ridder, D., Teusink, B., 2009. Shifts in growth strategies FEMS Yeast Res. 15, fov049.
reflect tradeoffs in cellular economics. Mol. Syst. Biol. 5, 323. Saitua, F., Torres, P., Perez-Correa, J.R., Agosin, E., 2017. Dynamic genome-scale meta-
Moser, J.W., Prielhofer, R., Gerner, S.M., Graf, A.B., Wilson, I.B., Mattanovich, D., bolic modeling of the yeast Pichia pastoris. BMC Syst. Biol. 11, 27.
Dragosits, M., 2017. Implications of evolutionary engineering for growth and re- Sánchez, B.J., Pérez-Correa, J.R., Agosin, E., 2014. Construction of robust dynamic
combinant protein production in methanol-based growth media in the yeast Pichia genome-scale metabolic model structures of Saccharomyces cerevisiae through itera-
pastoris. Microb. Cell Fact. 16, 49. tive re-parameterization. Metab. Eng. 25, 159–173.
Näätsaari, L., Mistlberger, B., Ruth, C., Hajek, T., Hartner, F.S., Glieder, A., 2012. Deletion Sánchez, B.J., Zhang, C., Nilsson, A., Lahtvee, P.J., Kerkhoven, E.J., Nielsen, J., 2017.
of the Pichia pastoris KU70 homologue facilitates platform strain generation for gene Improving the phenotype predictions of a yeast genome-scale metabolic model by
expression and synthetic biology. PLoS One 7, e39720. incorporating enzymatic constraints. Mol. Syst. Biol. 13, 935.
Naumov, G.I., Naumova, E.S., Tyurin, O.V., Kozlov, D.G., 2013. Komagataella kurtzmanii Sarkari, P., Marx, H., Blumhoff, M.L., Mattanovich, D., Sauer, M., Steiger, M.G., 2017. An
sp. nov., a new sibling species of Komagataella (Pichia) pastoris based on multigene efficient tool for metabolic pathway construction and gene integration for Aspergillus
sequence analysis. Antonie Van Leeuwenhoek 104, 339–347. niger. Bioresour. Technol. 245, 1327–1333.
Nett, J.H., Gerngross, T.U., 2003. Cloning and disruption of the PpURA5 gene and con- Sauer, U., 2006. Metabolic networks in motion: 13C-based flux analysis. Mol. Syst. Biol.
struction of a set of integration vectors for the stable genetic modification of Pichia 2, 62.
pastoris. Yeast 20, 1279–1290. Schreiber, C., Muller, H., Birrenbach, O., Klein, M., Heerd, D., Weidner, T., Salzig, D.,
Neubauer, S., Haberhauer-Troyer, C., Klavins, K., Russmayer, H., Steiger, M.G., Gasser, B., Czermak, P., 2017. A high-throughput expression screening platform to optimize the
Sauer, M., Mattanovich, D., Hann, S., Koellensperger, G., 2012. U13C cell extract of production of antimicrobial peptides. Microb. Cell Fact. 16, 29.
Pichia pastoris–a powerful tool for evaluation of sample preparation in metabolomics. Schwarzhans, J.P., Wibberg, D., Winkler, A., Luttermann, T., Kalinowski, J., Friehs, K.,
J. Sep. Sci. 35, 3091–3105. 2016. Integration event induced changes in recombinant protein productivity in
Nie, Y., Huang, M., Lu, J., Qian, J., Lin, W., Chu, J., Zhuang, Y., Zhang, S., 2014. Impacts Pichia pastoris discovered by whole genome sequencing and derived vector optimi-
of high beta-galactosidase expression on central metabolism of recombinant Pichia zation. Microb. Cell Fact. 15, 84.
pastoris GS115 using glucose as sole carbon source via 13C metabolic flux analysis. J. Schwarzhans, J.P., Luttermann, T., Geier, M., Kalinowski, J., Friehs, K., 2017a. Towards
Biotechnol. 187, 124–134. systems metabolic engineering in Pichia pastoris. Biotechnol. Adv. 35, 681–710.
Nielsen, J., 1998. Metabolic engineering: techniques for analysis of targets for genetic Schwarzhans, J.P., Luttermann, T., Wibberg, D., Winkler, A., Hubner, W., Huser, T.,

13
D.A. Peña et al. Metabolic Engineering xxx (xxxx) xxx–xxx

Kalinowski, J., Friehs, K., 2017b. A mitochondrial autonomously replicating se- 5, 172–186.
quence from Pichia pastoris for uniform high level recombinant protein production. Vogl, T., Gebbie, L., Palfreyman, R.W., Speight, R., 2018a. Effect of plasmid design and
Front Microbiol. 8, 780. type of integration event on recombinant protein expression in. Appl. Environ.
Shao, Z., Zhao, H., 2009. DNA assembler, an in vivo genetic method for rapid construction Microbiol. http://dx.doi.org/10.1128/AEM.02712-1.
of biochemical pathways. Nucleic Acids Res. 37, e16. Vogl, T., Sturmberger, L., Fauland, P.C., Hyden, P., Fischer, J.E., Schmid, C., Thallinger,
Shibui, T., Hara, H., 2017. A new type of gene-disruption cassette with a rescue gene for G.G., Geier, M., Glieder, A., 2018b. Methanol independent induction in Pichia pastoris
Pichia pastoris. Biotechnol. Prog. 33, 1201–1208. by simple derepressed overexpression of single transcription factors. Biotechnol.
Shin, S.K., Hyeon, J.E., Kim, Y.I., Kang, D.H., Kim, S.W., Park, C., Han, S.O., 2015. Bioeng. 115, 1037–1050.
Enhanced hydrolysis of lignocellulosic biomass: bi-functional enzyme complexes Vos, T., Hakkaart, X., van Maris, A., Pronk, J., Daran-Lapujade, P., 2016. Saccharomyces
expressed in Pichia pastoris improve bioethanol production from Miscanthus sinensis. cerevisiae is extremely robust and maintains high metabolic capacity at aerobic near
Biotechnol. J. 10, 1912–1919. zero-growth rates. Microb. Cell Fact. 15, 111.
Singh, V., Braddick, D., Dhar, P.K., 2017. Exploring the potential of genome editing Wagner, J.M., Alper, H.S., 2016. Synthetic biology and molecular genetics in non-con-
CRISPR-Cas9 technology. Gene 599, 1–18. ventional yeasts: current tools and future advances. Fungal Genet. Biol. 89, 126–136.
Siripong, W., Wolf, P., Kusumoputri, T.P., Downes, J.J., Kocharin, K., Tanapongpipat, S., Wang, J., Wang, X., Shi, L., Qi, F., Zhang, P., Zhang, Y., Zhou, X., Song, Z., Cai, M., 2017.
Runguphan, W., 2018. Metabolic engineering of Pichia pastoris for production of Methanol-independent protein expression by AOX1 promoter with trans-acting ele-
isobutanol and isobutyl acetate. Biotechnol. Biofuels 11, 1. ments engineering and glucose-glycerol-shift induction in Pichia pastoris. Sci. Rep. 7,
Sohn, S.B., Graf, A.B., Kim, T.Y., Gasser, B., Maurer, M., Ferrer, P., Mattanovich, D., Lee, 41850.
S.Y., 2010. Genome-scale metabolic model of methylotrophic yeast Pichia pastoris and Wegner, G.H., Harder, W., 1987. Methylotrophic yeasts–1986. Antonie Van Leeuwenhoek
its use for in silico analysis of heterologous protein production. Biotechnol. J. 5, 53, 29–36.
705–715. Wei, L., Wang, Z., Zhang, G., Ye, B., 2017. Characterization of terminators in
Solà, A., Maaheimo, H., Ylönen, K., Ferrer, P., Szyperski, T., 2004. Amino acid bio- Saccharomyces cerevisiae and an exploration of factors affecting their strength.
synthesis and metabolic flux profiling of Pichia pastoris. Eur. J. Biochem. 271, ChemBioChem 18, 2422–2427.
2462–2470. Weninger, A., Glieder, A., Vogl, T., 2015. A toolbox of endogenous and heterologous
Solà, A., Jouhten, P., Maaheimo, H., Sanchez-Ferrando, F., Szyperski, T., Ferrer, P., 2007. nuclear localization sequences for the methylotrophic yeast Pichia pastoris. FEMS
Metabolic flux profiling of Pichia pastoris grown on glycerol/methanol mixtures in Yeast Res. 15, fov082.
chemostat cultures at low and high dilution rates. Microbiology 153, 281–290. Weninger, A., Hatzl, A.M., Schmid, C., Vogl, T., Glieder, A., 2016. Combinatorial opti-
Sreekrishna, K., Tschopp, J.F., Fuke, M., 1987. Invertase gene (SUC2) of Saccharomyces mization of CRISPR/Cas9 expression enables precision genome engineering in the
cerevisiae as a dominant marker for transformation of Pichia pastoris. Gene 59, methylotrophic yeast Pichia pastoris. J. Biotechnol. 235, 139–149.
115–125. Weninger, A., Fischer, J., Raschmanova, H., Kniely, C., Vogl, T., Glieder, A., 2018.
Sturmberger, L., Chappell, T., Geier, M., Krainer, F., Day, K.J., Vide, U., Trstenjak, S., Expanding the CRISPR/Cas9 toolkit for Pichia pastoris with efficient donor integration
Schiefer, A., Richardson, T., Soriaga, L., Darnhofer, B., Birner-Gruenberger, R., Glick, and alternative resistance markers. J. Cell. Biochem. 119, 3183–3198.
B.S., Tolstorukov, I., Cregg, J., Madden, K., Glieder, A., 2016. Refined Pichia pastoris Wishart, D.S., 2007. Current progress in computational metabolomics. Brief. Bioinform. 8,
reference genome sequence. J. Biotechnol. 235, 121–131. 279–293.
Swinnen, S., Thevelein, J.M., Nevoigt, E., 2012. Genetic mapping of quantitative phe- Wriessnegger, T., Augustin, P., Engleder, M., Leitner, E., Muller, M., Kaluzna, I.,
notypic traits in Saccharomyces cerevisiae. FEMS Yeast Res. 12, 215–227. Schurmann, M., Mink, D., Zellnig, G., Schwab, H., Pichler, H., 2014. Production of
Theron, C.W., Berrios, J., Delvigne, F., Fickers, P., 2018. Integrating metabolic modeling the sesquiterpenoid (+)-nootkatone by metabolic engineering of Pichia pastoris.
and population heterogeneity analysis into optimizing recombinant protein produc- Metab. Eng. 24, 18–29.
tion by Komagataella (Pichia) pastoris. Appl. Microbiol. Biotechnol. 102, 63–80. Wu, G., Yan, Q., Jones, J.A., Tang, Y.J., Fong, S.S., Koffas, M.A.G., 2016. Metabolic
Tomas-Gamisans, M., Ferrer, P., Albiol, J., 2016. Integration and validation of the burden: cornerstones in synthetic biology and metabolic engineering applications.
genome-scale metabolic models of Pichia pastoris: a comprehensive update of protein Trends Biotechnol. 34, 652–664.
glycosylation pathways, lipid and energy metabolism. PLoS One 11, e0148031. Wu, S., Letchworth, G.J., 2004. High efficiency transformation by electroporation of
Tomàs-Gamisans, M., Ferrer, P., Albiol, J., 2018. Fine-tuning the P. pastoris iMT1026 Pichia pastoris pretreated with lithium acetate and dithiothreitol. Biotechniques 36,
genome-scale metabolic model for improved prediction of growth on methanol or 152–154.
glycerol as sole carbon sources. Microb. Biotechnol. 11, 224–237. Xue, Y., Kong, C., Shen, W., Bai, C., Ren, Y., Zhou, X., Zhang, Y., Cai, M., 2017.
Tredwell, G.D., Edwards-Jones, B., Leak, D.J., Bundy, J.G., 2011. The development of Methylotrophic yeast Pichia pastoris as a chassis organism for polyketide synthesis via
metabolomic sampling procedures for Pichia pastoris, and baseline metabolome data. the full citrinin biosynthetic pathway. J. Biotechnol. 242, 64–72.
PLoS One 6, e16286. Yamada, Y., Matsuda, M., Maeda, K., Mikata, K., 1995. The phylogenetic relationships of
Tredwell, G.D., Aw, R., Edwards-Jones, B., Leak, D.J., Bundy, J.G., 2017. Rapid screening methanol-assimilating yeasts based on the partial sequences of 18S and 26S ribo-
of cellular stress responses in recombinant Pichia pastoris strains using metabolite somal RNAs: the proposal of Komagataella gen. nov. (Saccharomycetaceae). Biosci.
profiling. J. Ind. Microbiol. Biotechnol 44, 413–417. Biotechnol. Biochem. 59, 439–444.
Tsakraklides, V., Brevnova, E., Stephanopoulos, G., Shaw, A.J., 2015. Improved gene Yamanishi, M., Ito, Y., Kintaka, R., Imamura, C., Katahira, S., Ikeuchi, A., Moriya, H.,
targeting through cell cycle synchronization. PLoS One 10, e0133434. Matsuyama, T., 2013. A genome-wide activity assessment of terminator regions in
Valli, M., Tatto, N.E., Peymann, A., Gruber, C., Landes, N., Ekker, H., Thallinger, G.G., Saccharomyces cerevisiae provides a ″terminatome″ toolbox. ACS Synth. Biol. 2,
Mattanovich, D., Gasser, B., Graf, A.B., 2016. Curation of the genome annotation of 337–347.
Pichia pastoris (Komagataella phaffii) CBS7435 from gene level to protein function. Yang, J., Cai, H., Liu, J., Zeng, M., Chen, J., Cheng, Q., Zhang, L., 2018. Controlling AOX1
FEMS Yeast Res. 16, fow051. promoter strength in Pichia pastoris by manipulating poly (dA:dT) tracts. Sci. Rep. 8,
van der Klei, I.J., Yurimoto, H., Sakai, Y., Veenhuis, M., 2006. The significance of per- 1401.
oxisomes in methanol metabolism in methylotrophic yeast. Biochim. Biophys. Acta Yang, Z., Zhang, Z., 2018. Engineering strategies for enhanced production of protein and
1763, 1453–1462. bio-products in Pichia pastoris: a review. Biotechnol. Adv. 36, 182–195.
Vasilakou, E., Machado, D., Theorell, A., Rocha, I., Nöh, K., Oldiges, M., Wahl, S.A., 2016. Ye, J., Ly, J., Watts, K., Hsu, A., Walker, A., McLaughlin, K., Berdichevsky, M., Prinz, B.,
Current state and challenges for dynamic metabolic modeling. Curr. Opin. Microbiol. Sean Kersey, D., d'Anjou, M., Pollard, D., Potgieter, T., 2011. Optimization of a
33, 97–104. glycoengineered Pichia pastoris cultivation process for commercial antibody produc-
Velagapudi, V.R., Wittmann, C., Schneider, K., Heinzle, E., 2007. Metabolic flux screening tion. Biotechnol. Prog. 27, 1744–1750.
of Saccharomyces cerevisiae single knockout strains on glucose and galactose supports Ye, R., Huang, M., Lu, H., Qian, J., Lin, W., Chu, J., Zhuang, Y., Zhang, S., 2017.
elucidation of gene function. J. Biotechnol. 132, 395–404. Comprehensive reconstruction and evaluation of Pichia pastoris genome-scale meta-
Vervecken, W., Kaigorodov, V., Callewaert, N., Geysens, S., De Vusser, K., Contreras, R., bolic model that accounts for 1243 ORFs. Bioresour. Bioprocess. 4, 22.
2004. In vivo synthesis of mammalian-like, hybrid-type N-glycans in Pichia pastoris. Zahrl, R.J., Pena, D.A., Mattanovich, D., Gasser, B., 2017. Systems biotechnology for
Appl. Environ. Microbiol. 70, 2639–2646. protein production in Pichia pastoris. FEMS Yeast Res. 17, fox068.
Vogl, T., Glieder, A., 2013. Regulation of Pichia pastoris promoters and its consequences Zamboni, N., Fendt, S.M., Rühl, M., Sauer, U., 2009. 13C-based metabolic flux analysis.
for protein production. New Biotechnol. 30, 385–404. Nat. Protoc. 4, 878–892.
Vogl, T., Ruth, C., Pitzer, J., Kickenweiz, T., Glieder, A., 2014. Synthetic core promoters Zhang, M., Yu, X.W., Xu, Y., Jouhten, P., Swapna, G.V.T., Glaser, R.W., Hunt, J.F.,
for Pichia pastoris. ACS Synth. Biol. 3, 188–191. Montelione, G.T., Maaheimo, H., Szyperski, T., 2017. 13C metabolic flux profiling of
Vogl, T., Ahmad, M., Krainer, F.W., Schwab, H., Glieder, A., 2015. Restriction site free Pichia pastoris grown in aerobic batch cultures on glucose revealed high relative
cloning (RSFC) plasmid family for seamless, sequence independent cloning in Pichia anabolic use of TCA cycle and limited incorporation of provided precursors of
pastoris. Microb. Cell Fact. 14, 103. branched-chain amino acids. FEBS J. 284, 3100–3113.
Vogl, T., Sturmberger, L., Kickenweiz, T., Wasmayer, R., Schmid, C., Hatzl, A.M., Zhao, C., Gao, X., Liu, X., Wang, Y., Yang, S., Wang, F., Ren, Y., 2016. Enhancing bio-
Gerstmann, M.A., Pitzer, J., Wagner, M., Thallinger, G.G., Geier, M., Glieder, A., synthesis of a ginsenoside precursor by self-assembly of two key enzymes in Pichia
2016. A toolbox of diverse promoters related to methanol utilization: functionally pastoris. J. Agric. Food Chem. 64, 3380–3385.
verified parts for heterologous pathway expression in Pichia pastoris. ACS Synth. Biol.

14

Vous aimerez peut-être aussi