You are on page 1of 14

Appl Microbiol Biotechnol (2011) 90:1193–1206

DOI 10.1007/s00253-011-3212-8

MINI-REVIEW

An overview of lipid metabolism in yeasts and its impact


on biotechnological processes
Athanasios Beopoulos & Jean-Marc Nicaud &
Claude Gaillardin

Received: 25 January 2011 / Accepted: 25 January 2011 / Published online: 31 March 2011
# Springer-Verlag 2011

Abstract High energy prices, depletion of crude oil Introduction


supplies, and price imbalance created by the increasing
demand of plant oils or animal fat for biodiesel and specific The role of lipids, their precursors, and their derivatives in
lipid derivatives such as lubricants, adhesives, and plastics eukaryotic cells is being increasingly recognized. The
have given rise to heated debates on land-use practices and emerging field of lipidomics, driven by the progress in
to environmental concerns about oil production strategies. genetic engineering and amplified by the technological
However, commercialization of microbial oils with similar advances in lipid analysis, is attracting more and more
composition and energy value to plant and animal oils interest for the study of lipid metabolism in various
could have many advantages, such as being non- organisms. Among them, the yeast Saccharomyces cerevi-
competitive with food, having shorter process cycle and siae offers a number of advantages for lipidomics due to the
being independent of season and climate factors. This high accessibility of its molecular and classical genetics, the
review focuses on the ongoing research on different ease of cultivation, and its short generation time. As a
oleaginous yeasts producing high added value lipids and consequence, the increasing range of available yeast
on the prospects of such microbial oils to be used in mutants offers great opportunities to perturb or to modulate
different biotechnological processes and applications. It lipid metabolism to study the effects produced at the
covers the basic biochemical mechanisms of lipid synthesis molecular and/or cellular level (Nielsen 2009).
and accumulation in these organisms, along with the latest Besides S. cerevisiae, which has long been used for
insights on the metabolic processes involved. The key fermentation of food and beverages, several other species
elements of lipid accumulation, the mechanisms suspected are nowadays widely considered for industrial production
to confer the oleaginous character of the cell, and the of chemicals, pharmaceuticals, and proteins. Interestingly,
potential metabolic routes enhancing lipid production are while the majority of yeasts are potentially well adapted to
also extensively discussed. these procedures, there is only a small class among them
which has the capacity to store lipids to over 20% of the
Keywords Yeast . Lipid metabolism . Bio-oils . biomass. These yeasts, called oleaginous, are generally
Biotechnology . Genetic engineering non-pathogenic and are able to synthesize and accumulate
lipids with a composition similar to vegetable oils and fats.
As environmental protection issues and depletion of crude
A. Beopoulos : J.-M. Nicaud : C. Gaillardin (*) oil supplies are becoming the overriding challenges faced
AgroParisTech, UMR1319, Micalis,
by oil industries, it comes not as a surprise to see
Centre de Biotechnologie Agro-Industrielle,
78850 Thiverval-Grignon, France biotechnological companies increasingly investing on the
e-mail: claude.gaillardin@grignon.inra.fr development of first- and second-generation biofuels from
plants and crop residues, respectively. Biodiesel, by means
A. Beopoulos : J.-M. Nicaud : C. Gaillardin
of transesterified fatty acids (fatty acid methyl esters—
INRA, UMR1319, Micalis,
Centre de Biotechnologie Agro-Industrielle, FAME) is biodegradable, non-toxic, and essentially free of
78850 Thiverval-Grignon, France sulfur and aromatic components. However, more than 95%
1194 Appl Microbiol Biotechnol (2011) 90:1193–1206

of the world’s biodiesel is derived from edible vegetable and a small fraction of steryl esters (SE), collectively called
oils, increasing the worldwide demand for vegetable oil neutral lipids. These storage lipids accumulate in a
production (Gui et al. 2008). The most commonly used specialized compartment of the cell known as the lipid
vegetable oils for biodiesel production are palm, soybean, body (LB). The latter consists of a lipid core encased in a
rapeseed, and tung oil. It is estimated that by the end of this phospholipid monolayer within which many proteins with
year world biodiesel production will reach 11.1 million diverse functions are embedded (Zweytick et al. 2000;
tons/year with its production capacity rising to 32.6 million Brown 2001; Fujimoto et al. 2008). The long prevailing
tons/year, following an annual growth rate of 40% since idea that LBs are simply storage compartments has been
2002 (Thurmond 2008). As a consequence, from 2005 to however challenged in higher eukaryotic cells as several
the present day, prices of a number of vegetable oils have LB proteins were shown to play key roles in lipid storage,
largely increased due to biofuel demand. lipid biosynthesis, metabolism, degradation, and substrate
Today, however, it seems possible to consider micro- trafficking (Brown 2001; Beckman 2006). In S. cerevisiae,
organisms as factories for the production of molecules with LB are reported to contain mainly if not exclusively
equivalent or improved properties compared to those enzymes involved in neutral lipid catabolism, whereas
derived from fossil oils. The production line of microbial biosynthetic enzymes are deemed to be present exclusively
oils from plant biomass may not compete with food in the ER (Czabany et al. 2007). In the oleaginous yeast
production if waste products such as lignocelluloses are Yarrowia lipolytica, analysis of LB proteins evidenced
used as substrates. It could eventually present other anabolic and catabolic enzymes, as well as proteins
advantages compared to plants, such as shorter process controlling vesicle trafficking, a situation clearly reminis-
cycle, independence of season and climate, and easier cent of that described in adipocyte cells (Athenstaedt and
scale-up. Furthermore, the emergence of new downstream Daum 2006). Moreover, it has recently been recognized
processing techniques results in an optimistic forecast for that free fatty acids (FFA) accumulate in LBs as well, and a
the commercialization of yeast–lipid products in the future. lipid-binding protein from the oleaginous yeast Y. lipolytica
An innovative approach towards this end uses white LB has been involved in lipid trafficking between the
biotechnology and metabolic engineering of microorgan- cytoplasm and LB (Mlickova et al. 2004b; Athenstaedt et
isms for both the production of biofuels and non-fuel-oil- al. 2006; Beckman 2006; Ferreyra et al. 2006).
derived products such as surfactants, solvents, paints, On another hand, lipid content and profile of LBs differ
waxes, plastics, and lubricants to name a few. It is between species. Table 1 illustrates the wide range of fatty
important to state, however, that only high valued oils have acids (FA) found in yeasts together with the extent of lipid
any chance of being produced by such biotechnological accumulation of the species undergoing fermentation on
means as it is currently unrealistic to expect microorgan- glucose containing medium. For instance, Cryptococcus
isms to produce main commodity oils and fats as cheaply as curvatus and Cryptococcus albidus may accumulate lipids
do plant and animal sources. to equivalent levels (58% and 65%, respectively), but their
Among microorganisms, yeasts, being unicellular, de- fatty acid profiles vary: C. curvatus accumulates significant
void of endotoxins, amenable to genetic improvement and amounts of palmitic acid (C16:0), whereas oleic acid
suitable for large-scale fermentation, are particularly attrac- (C18:1, n−9) is preferentially accumulated by C. albidus.
tive for the development of biotechnological approaches. In contrast, total lipid content varies significantly between
More precisely, oleaginous yeasts seem to be the best different Rhodotorula species, but fatty acid composition
candidates for this purpose. remains similar. It can be acknowledged, however, that the
main fatty acids produced by oleaginous yeasts are myristic
(C14:0), palmitic (C16:0), palmitoleic (C16:1, n−7), stearic
Oleaginous yeasts (C18:0), oleic (C18:1, n−9), linoleic (C18:2, n−6), and
linolenic acid (C18:3). It has been suggested that such yeast
Fewer than 30 of the about 600 yeast species are known to oils could be used as oil feedstocks for biodiesel production
be oleaginous (Rattray 1988; Ratledge 1989, 1994). that require C16–C18 fatty acids (Steen et al. 2010).
Oleaginous yeasts are typically found, but not exclusively, In all cases, process development will require further
in genera such as Candida, Cryptococcus, Rhodotorula, metabolic engineering of the strains to increase as much as
Rhizopus, Trichosporon, Lipomyces, and Yarrowia (Ratlege possible lipid yields and production rates on cheap raw
and Tan 1990). On average, these yeasts accumulate lipids materials as carbon sources. In order to prepare the stage
to levels corresponding to 40% of their biomass. However, for these developments, we will first provide a description
under conditions of nutrient limitation (see below), they of yeast lipid metabolism, assessing the key elements of
may accumulate lipids to levels exceeding 70% of their lipid accumulation and the mechanisms suspected to confer
biomass. They consist of 80–90% triacylglycerols (TAG) the oleaginous character to these cells. We will frequently
Appl Microbiol Biotechnol (2011) 90:1193–1206 1195

Table 1 Lipid accumulation and fatty acid profiles of selected oleaginous yeasts grown on glucose containing medium (data adapted from
Ratledge and Wynn 2002 and complemented by our own experiences on Y. lipolytica)

Species Lipid accumulation (% D.W.) Major fatty acid residues (% w/w)

C16:0 C16:1 C18:0 C18:1 C18:2 C18:3

Cryptococcus curvatus 58 25 Trace 10 57 7 0


Cryptococcus albidus 65 12 1 3 73 12 0
Candida sp 107 42 44 5 8 31 9 1
Lipomyces starkeyi 63 34 6 5 51 3 0
Rhodotorula glutinis 72 37 1 3 47 8 0
Rhodotorula graminis 36 30 2 12 36 15 4
Rhizopus arrhizus 57 18 0 6 22 10 12
Trichosporon pullulans 65 15 0 2 57 24 1
Yarrowia lipolytica 36 11 6 1 28 51 1

make reference to the non-oleaginous yeast S. cerevisiae recent data on the biochemistry and regulation of fatty acid
which remains the best studied model for lipid metabolism, synthesis and elongation in S. cerevisiae have been
and to Y. lipolytica, an oleaginous yeast which is genetically reviewed by Tehlivets et al. (2007).
tractable contrary to most other oleaginous species. How-
ever, other yeast models and species will be addressed too Fatty acid synthesis in yeasts
whenever data are available.
De novo fatty acid synthesis in yeast is carried out by the
fatty acid synthetase (FAS) enzymatic complex in the
Overview of yeast lipid metabolism cytosol and requires the constant supply of acetyl-CoA
and malonyl-CoA. The former is the initial biosynthetic
An intriguing question that comes to mind when looking at unit and the latter the elongation unit, providing two
the different lipid accumulation rates among yeasts, carbons at each step of the growing fatty acid chain. The
whether oleaginous or not, concerns the origin of these origin of acetyl-CoA, however, differs in oleaginous and
differences. Why do some microorganisms accumulate non-oleaginous species. Non-oleaginous yeasts produce
lipids and others do not, and why do so vast fluctuations acetyl-CoA from glycolysis of fermentable sugars by
in lipid content exist between oleaginous yeasts? breaking down pyruvate in the mitochondrion and by the
At first, we should differentiate the processes of lipid cytosolic pyruvate–acetaldehyde–acetate pathway. In ole-
synthesis and lipid accumulation. De novo lipid synthesis aginous microorganisms, an additional source of acetyl-
provides for the synthesis of fatty acid precursors, such as CoA exists: its precursor is the excess of citrate exported
acetyl- and malonyl-CoA and for their subsequent elonga- from the Krebs cycle out of the mitochondrion, via the
tion to an intermediate level (C14–C16), depending on the malate/citrate shuttle. Citrate is then cleaved within the
enzymatic arsenal of each organism. These precursors then cytosol by ATP citrate lyase (ACL), an enzyme present in
feed into the lipid storage pathway, where the fatty acyl- all oleaginous microorganisms studied so far (Ratledge
CoAs, assemble to form triacylglycerols (TAG) and steryl 2004):
esters (SE). The neutral lipids are then stored inside the LB
to serve as energy reserves. In the presence of external Citrate þ CoA þ ATP ! acetyl ⁃⁃ CoA þ oxaloacetate
sources of fatty acids, oils, or occasionally alkanes, yeasts þ ADP þ Pi
proceed to the incorporation of these substrates and their
subsequent modification for storage and accumulation. This It is thus quite likely that ACL is one of the enzymes
pathway, also known as the ex novo pathway, may require contributing to the oleaginous character of the cell, even if
the hydrolysis of the hydrophobic substrate (HS) and is some non-oleaginous yeasts exhibit ACL activity. Further-
followed by the incorporation/transport of the released fatty more, this enzyme appears to be regulated by the presence
acids in the form of CoA-thioesters which feed into the of exogenous fatty acids. In a study of various oleaginous
storage lipid pathway to generate neutral lipids. An early yeast species (Holdsworth et al. 1988), there was a
review on fatty acid biosynthetic pathways in oleaginous considerable decrease in ACL activity, in all species
yeasts can be found in Ratledge and Wynn (2002), while examined, as soon as yeasts utilized endogenous or
1196 Appl Microbiol Biotechnol (2011) 90:1193–1206

exogenous lipids. ACL is made up of two subunits, elongation (Ratledge 2004). This was suggested by studies
encoded by the ACL1 and ACL2 genes and requires for its on Mucor circinelloides and Aspergillus nidulans, where
activation an ammonium ion, which may be scarce in the inhibition or mutation of ME resulted in a loss of lipid
absence of nitrogen, or under high C/N conditions, accumulation (Wynn et al. 1997). Reciprocally, the over-
common approaches used in order to induce lipid accumu- expression of the NADP(+) ME of M. circinelloides
lation (see below) (Botham and Ratledge 1979; Ratledge resulted in a 2.5-fold increase in lipid accumulation (Zhang
and Wynn 2002). This probably accounts for the difficulty et al. 2007). This, however, would suggest that both a
to maintain de novo FA synthesis when yeasts are fed mitochondrial and a cytoplasmic form of ME should exist
exogenous lipids, even if the C/N ratio is kept high. in yeasts, as in Schizosaccharomyces pombe and most
Concerning malonyl-CoA, it is generated by the acetyl- eukaryotic cells, but not in S. cerevisiae where a single gene
CoA carboxylase (ACC) enzyme by the condensation of an encoding a mitochondrial product has been identified
acetyl-CoA unit with a bicarbonate anion: (Boles et al. 1998). A cytoplasmic form has indeed been
reported in Lipomyces starkei, but it was shown to strongly
acetyl ⁃⁃ CoA þ HCO3  þ ATP ! malonyl ⁃⁃ CoA prefer NAD+ over NADP+ (Tang et al. 2010). Furthermore,
þ ADP þ Pi a single mitochondrial form is predicted to exist in Y.
lipolytica (YALI0E18634g), albeit its overexpression did
The ACC enzyme appears to be crucial for de novo FA not affect lipid accumulation (our unpublished data). Thus,
synthesis as mutants invalidated for ACC are either the existence of such a ME–FAS–ACL complex does not
incapable of synthesizing their own FA (FA auxotrophs) seem to characterize all oleaginous yeasts and ME activity
or may harbor low levels of residual ACC activity or NADPH concentration and origin may not be the
(Tehlivets et al. 2007). limiting factors for lipid accumulation in oleaginous yeasts.
The FAS biosynthetic complex in yeasts is composed of It should be stressed at this point that, besides the
two subunits, Fas1 (beta subunit) and Fas2 (alpha subunit), classical (or FAS I) cytoplasmic pathway of FA synthesis
which are organized as a hexameric α6β6 complex. mediated by the Fas1/Fas2 complex, a mitochondrial
Comprehensive reviews on fatty acid synthetases have pathway has been evidenced in S. cerevisae and in most
recently been published by Schweizer and Hofmann (2004) eukaryotes, similar to the bacterial FAS II pathway (for a
and Tehlivets et al. (2007). Transcription of the FAS1, FAS2, recent review, the reader is referred to Hiltunen et al. 2009).
and ACC1 are all decreased in the presence of fatty acids. If It relies on an acyl-carrier protein (ACP) and produces
FAS2 is overexpressed in cells, the expression of FAS1 and octanoyl-ACP (a precursor of lipoic acid) and possibly
ACC1 is also increased, indicating coordinate regulation of longer FA. Its inactivation in S. cerevisiae results in
these three genes. In addition to fatty acid repression, these mitochondrial defects (impairment of mRNA maturation,
three genes are coordinately regulated by an inositol/ cytochrome losses, and respiratory deficiency), but its role
choline response element, along with a number of genes in other yeasts has hitherto not been investigated.
involved in phospholipid biosynthesis (Chirala 1992;
Hasslacher et al. 1993). It is thus likely that one of these Fatty acid modification reactions
three enzymes acts as a limiting factor in lipid accumula-
tion. In keeping with this idea, a 40% increase in the total The FAS end product depends on yeast species, but varies
fatty acid content of the non-oleaginous yeast Hansenula between 14 and 16 carbon saturated fatty acids. Further
polymorpha was achieved by overexpressing the ACC1 elongation or desaturation takes place in the ER by the
from the oleaginous fungus Mucor rouxii (Ruenwai et al. respective elongases and/or desaturases of the species.
2009). The cyclic series of elongation reactions, reminiscent of
For each step of the carbon chain elongation reaction, de novo lipid synthesis, require malonyl-CoA, provided by
FAS requires two molecules of NADPH. Cellular NADPH the ACC. All elongation enzymes identified in yeasts
can be either produced by the malic enzyme (malate (ELO1, ELO2, ELO3, TSC13, PHS1, and YBR159w) are
dehydrogenase carboxylase) by the reaction: localized in the endoplasmic reticulum (ER), where they are
presumably organized in a complex (Kohlwein et al. 2001;
malate þ NADPþ ! pyruvate þ NADPH
Denic and Weissman 2007; Tehlivets et al. 2007; Kihara et
or in the cytoplasm by the pentose phosphate pathway al. 2008). The major very long-chained fatty acid (VLCFA)
through glucose-6-dehydrogenase, 6-phosphogluconate de- species in yeast is C26, predominantly present in an amide
hydrogenase, and NADPH isocitrate dehydrogenase. How- linkage of the ceramide backbone of sphingolipids.
ever, malic enzyme (ME) has been found in most Desaturases are hydrophobic, membrane-bound proteins
oleaginous microorganisms and has been suggested to form endowed with at least three separate functions (cytochrome
a complex with ACL and FAS so as to facilitate FA b5 reductase, cytochrome b5 oxidase, dehydrogenase) to
Appl Microbiol Biotechnol (2011) 90:1193–1206 1197

yield the final activity. Their substrate consists of either AMP-binding protein with a great similarity to acyl-CoA
acyl-CoA or acyl residues attached on the head of a synthetases, is also believed to participate in the import of
phospholipid, indicating that a number of specialized FA. In S. cerevisiae, it is localized in the peripheral
acyltransferases and phosphatidylcholine metabolic membrane and matrix (Biobel and Erdmann 1996). How-
enzymes likely participate in these reactions (Dahlqvist et ever, it is not required for growth under conditions in which
al. 2000). The most commonly found desaturases in yeasts the peroxisomal β-oxidation of fatty acids provides the
are the Δ9 and Δ12 desaturase and, in some species such as carbon source (Hiltunen et al. 2003). Recently another
Rhodotorula glacialis and Debaryomyces hansenii, the Δ15 protein, Ypk1p, the yeast orthologue of the human serum
(both species) and Δ6 (R. glacialis) desaturases. The Δ9 and glucocorticoid-induced kinase Sgk1, was found to be
desaturase catalyses the insertion of the first double bond required for the efficient uptake of fatty acids in S.
into a saturated fatty acid (C16:0 and C18:0 produced by cerevisiae, and cells lacking Ypk1p fail to grow under fatty
FA neosynthesis) and is the only desaturase that acts on a acid auxotrophic conditions. Ypk1p is required for the
saturated acyl-CoA rather than a phospholipid. Mutants internalization step of endocytosis, suggesting that mem-
deficient in Δ9 activity are unable to grow in the absence of brane internalization is required for the uptake of fatty acids
exogenous unsaturated fatty acids (Goodrich-Tanrikulu et (Jacquier and Schneiter 2010). Once inside the cytoplasm,
al. 1994). The Δ12 desaturase catalyzes the insertion of the the FA is handled by several transporter systems such as the
second double bond (between carbons 12 and 13) into an fatty acid binding proteins (FABP), the ABC transporters,
oleic acid substrate to produce linoleic acid (C18:2, n−6). or the PXA and PEX11 systems (Theodoulou et al. 2006).
Further on, the Δ15 desaturase, if present, converts linoleic Different yeast species may have elaborated different
acid to γ-linolenic acid (C18:3, n−3). The Δ6 desaturase of systems, as for instance FABP are absent for S. cerevisiae
R. glacialis produces C18:3 n−6 from linoleic acid, or (Hiltunen et al. 2003), but very efficient in Y. lipolytica
C18:4 n−3 from γ-linolenic acid (Amaretti et al. 2010). (Dell'Angelica et al. 1992). The incorporated FA, in its
coenzyme A form, can then be directed either towards the
Incorporation of HS substrates—the ex novo accumulation neutral lipid storage pathway in an unchanged or modified
pathway form, or towards the β-oxidation pathway to be broken
down for energy production.
The elongation and desaturation reactions also target HS Alkane assimilation requires additional modifications in
substrates incorporated by the ex novo pathway. This order to serve for the synthesis of cellular compounds or for
pathway is more efficient in yeasts specialized in the FA biogenesis. Alkanes are classically considered to enter
degradation and hydrolysis of hydrophobic substrates (HS), directly in the cell and to then follow the monoterminal
notably the ascomycetous families of Endomycetaceae and oxidation pathway in the ER and the peroxisomes to be
Saccharomycetoideae. The most common species include converted into the corresponding fatty acids. However, a
Pichia jadinii, Candida rugosa, Y. lipolytica, and Torulop- recent analysis of Y. lipolytica mutants defective in ABC
sis colliculosa. The mechanisms elaborated by these species transporter genes suggested that such proteins are involved
include a surface-mediated transport, carried out by in the import or export of alkanes (Thevenieau 2006). The
surfactants such as lipases and emulsifiers, resulting in the oxidation reaction of the incorporated alkane is initiated by
reduction of the HS droplet size, and a direct interfacial a terminal hydroxylation carried out by a cytochrome P450-
transport which entails the formation of protrusions allow- dependent alkane monooxygenase system (AMOS, coded
ing HS droplet binding to the cell surface (Fickers et al. by the ALK genes), for which a 12 gene family has been
2005a; Beopoulos et al. 2009b). Yeasts like Y. lipolytica described in Y. lipolytica (Iida et al. 2000; Dujon et al.
which are specialized in HS utilization also underwent 2004). The fatty alcohol produced is then transformed into
amplification and successive evolution of the genes the corresponding aldehyde. This step is performed either
required for the utilization of HS of various chain length by a NAD+/NADP+-dependent fatty alcohol dehydrogenase
(Dujon et al. 2004). In addition, several membrane proteins (encoded by the ADH genes) or by a fatty alcohol oxidase
such as acyl-CoA synthetases have been proposed as (encoded by the FAO genes). Finally, the aldehyde is
possible fatty acid transporters or receptors, and have been converted into the corresponding FFA by a NAD+/NADP+-
shown to enhance fatty acid uptake into cells (DiRusso and dependent fatty aldehyde dehydrogenase (ALD genes).
Black 1999). This is the function of the FAT1/2 genes. FAT1 Nevertheless, at this point, a dicarboxylic acid (DCA) can
encodes an acyl-CoA synthetase required for both the be produced instead of a FFA by the intermediary of ω-
import of long-chain fatty acids and the activation of very oxidation carried out by a cytochrome P450-dependent
long-chain fatty acids (C20–C26). Disruption of FAT1 fatty acid ω-hydroxylase (diterminal oxidation or ω-
results in a decrease in the uptake of fatty acids (Faergeman oxidation) (Thevenieau et al. 2007). The fate of hydropho-
et al. 1997; Zou et al. 2002). FAT2, being a peroxisomal bic substrates is depicted in Fig. 1.
1198 Appl Microbiol Biotechnol (2011) 90:1193–1206

n-alkanes FFA TAG


protrusion LIP2
ABC1
P450
(AMOS)
FAT1/2
FFA-OH ER
FAO/ADH

cytosol
ALK1-12 aldehyde ALD
DCA FFA

ACL1/2 ER FAT1/2
TCA citrate acetyl-CoA
elongation/desaturation
ACC1/2 FAS1/2 acyl-CoA
NADP+ Sterol
MAE1?
acetyl-CoA malonyl-CoA
NADPH
ARE1

DGA1/2
SE lipid body
SCT1 SLC1 PAP1 LRO1
G3P LPA PA DAG TAG
TGL1
GUT2 GPD1
storage lipid synthesis Sterol
DHAP TGL2/3
mitochondrion
FAA1/4
acyl-CoA LC-FFA MC-FFA

PXA1/2
glucose fructose 1,6-bisP
FAA2
acyl-CoA FFA
POX1-6
MFE1
Carnitine shuttle POT1
or
Glyoxylate cycle acetyl-CoA
(pero-mito-cyto)
peroxisome

Fig. 1 Overview of lipid metabolism in yeasts with the corresponding alcohol dehydrogenase; ALK: cytochrome P450 oxidase; ARE:
gene names. Model created from the available data by Y. lipolytica. acyl-CoA:cholesterol acyltransferase; DGA: acyl-CoA:DAG acyl-
Abbreviations used for metabolic intermediates: DAG diacylglycerol, transferase; FAA: fatty acyl-CoA synthetase; FAD: fatty aldehyde
DCA dicarboxylic acid, DHAP dihydroxyacetone phosphate, FFA free dehydrogenase; FAO: fatty alcohol oxidase; FAS: fatty acid synthetase;
fatty acids, FFA-OH hydroxylated fatty acid, G3P glycerol-3- FAT: fatty acid import and activation; GUT: G-3-P dehydrogenase/
phosphate, LC-FFA long-chain fatty acids, LPA lysophosphatidic acid, glycerol kinase; LIP: lipase; LRO: phospholipid:diacylglycerol acyl-
MC-FFA medium-chain fatty acid, PA phosphatidic acid, TAG transferase; MAE: malic enzyme; MFE: multifunctional enzyme; PAP:
triacylglycerol, SE steryl ester, TCA tricarboxylic acid cycle. Gene phosphatidate phosphatase; POT: thiolase; POX: acyl-CoA oxidase;
names refer to following encoded enzymatic activities (see text for PXA: peroxisomal acyl-CoA transporter; TGL: triacylglycerol lipase;
details): ACC acetyl-CoA carboxylase; ACL: ATP citrate lyase; ADH: SCT: glycerol-3-phosphate acyltransferase; SLC: LPA acyltransferase

Neutral lipid formation TAG biosynthesis and homeostasis in S. cerevisiae, the


reader is referred to Czabany et al. (2007), Rajakumari et al.
The fatty acids produced by the de novo lipid synthesis or (2008), Kohlwein (2010), and Gaspar et al. (2011).
incorporated from the medium are then esterified either in At a branchpoint in neutral lipid synthesis, diacylgly-
the glycerol backbone or in a sterol molecule to form TAGs cerol (DAG) is formed: it is the direct precursor of TAG
and steryl esters (SE), respectively. The whole process is and phospholipids such as phosphatidylcholine and phos-
known as the storage lipid pathway, and its end products phatidylethanolamine. This molecule can be either pro-
form the neutral lipid fraction of the cell, packed inside the duced by the glycerol-3-phosphate pathway or by the
LB. The TAGs are very efficient molecules for energy monoacylglycerol (MAG) pathway. In this last one, DAG
storage as their prevalent hydrophobic properties allow is synthesized by direct acylation of MAG by an acyl-CoA:
tight packing inside the LB without affecting the polar diacylglycerol acyltransferase (Heier et al. 2010). Both
hydrophilic environment of the cell. For recent reviews on routes utilize fatty acyl-CoAs for the acylation of glycerol,
Appl Microbiol Biotechnol (2011) 90:1193–1206 1199

but the MAG pathway is not yet found in all yeast cells family and is the major player for TAG biosynthesis
(Coleman and Lee 2004). In the glycerol-3-phosphate (G-3- (Sandager et al. 2002). In both Y. lipolytica and S.
P) pathway, the sequential acylation of glycerol starts with cerevisiae, Dga1p are integral ER proteins and are more
the formation of G-3-P. This molecule is either derived active during stationary phase, although they are expressed
from glycerol via glycerol kinase (GUT1) or synthesized during all growth phases; in Y. lipolytica, Dga1p is one of
from dihydroxyacetone phosphate (DHAP) by G-3-P the LB proteins identified by Athenstaedt et al. (2006).
dehydrogenase (GPD1). This last reaction is reversible Dga1p accounts for 87% of DAG esterification activity in
and is catalyzed by an isoform of the G-3-P dehydrogenase S. cerevisiae (Sandager et al. 2002), but only for about 45%
(GUT2). Then, glycerol-3-phosphate acyltransferase in Y. lipolytica (Beopoulos et al., AEM submitted). The
(SCT1) generates lysophosphatidic acid (LPA), which, reason for these differences is that Y. lipolytica possesses a
however, can also be formed by reduction of DHAP by second gene coding for a DGAT acyltransferase, this time
an NADPH-dependent acyl-DHAP reductase (AYR1). The belonging in the DGAT1 family of DAG acyltransferases.
acylation of LPA is catalyzed by the LPA acyltransferase The protein, named Dga2p, accounts for about 30% of TAG
(SLC1) to yield phosphatidic acid (PA). After dephosphor- synthesis and appears to be more active during exponential
ylation of PA by phosphatidate phosphatase (PAP1), the growth phase. Members of the DGAT1 family are highly
DAG is formed. Interestingly, Slc1p and Sct1p have been similar to acyl-CoA:cholesterol acyltransferases (ACAT),
reported to have a dual localization in the ER and the LB in catalyzing sterol esterification in eukaryotes. In S. cerevi-
both S. cerevisiae and Y. lipolytica (Athenstaedt and Daum siae, there are two ACAT proteins, Are1p and Are2p,
2006). which are responsible for the whole process of sterol
Finally, the acylation of DAG is catalyzed either by an esterification but also able to exhibit minor DGAT activity.
acyl-CoA-dependent or independent reaction. The latter is In Y. lipolytica, however, only one gene belongs to the
carried out by a phospholipid:diacylglycerol acyltransferase ACAT family, YlARE1, and its product is incapable of
(PDAT), commonly known as Lro1p in yeasts, using the carrying out DAG esterification. For what concerns the Y.
sn-2 group of phospholipids as an acyl donor (Dahlqvist et lipolytica Dga2p, although it exhibits considerable similar-
al. 2000). However, although mammalian orthologues ity with YlAre1p, it is not involved in sterol esterification at
of PDAT proteins (lecithin:cholesterol acyltransferase all. As a likely consequence of these species differences,
family—LCAT) are able to synthesize sterols as well, in the neutral lipid composition of the two types of cells differ
yeasts such activity has not been observed. In S. cerevisiae, markedly: in S. cerevisiae, LBs contain equivalent propor-
Lro1p is localized in the ER, whereas in Y. lipolytica it tions of SE and TAG, while in Y. lipolytica SE represent
localizes to both the ER and the surface of LB (Athenstaedt only 5%, the rest being TAGs. Another interesting property
et al. 2006), playing a significant role in DAG acylation of Y. lipolytica is its ability to accumulate FFA as well.
during logarithmic growth in both organisms (Sandager et Nevertheless, the possibility that these molecules are
al. 2002). This, apart from a direct involvement of LBs in accommodated in the LB is still under investigation, but
neutral lipid synthesis, may also indicate a more efficient preliminary experimental data strongly suggest that this
TAG synthetic pathway in oleaginous yeasts. Unfortunate- may be the case (Beopoulos et al. 2008).
ly, localization data from other yeasts are currently not
available. Concerning the second, acyl-CoA-dependent Lipid catabolism
pathway, two main protein families with diacyl-glycerol
transferase (DGAT) activity have been identified in higher Neutral lipids stored in the LB can be mobilized to fulfill
eukaryotes and called DGAT1 and DGAT2. The DGAT upon demand the cell requirements for sterols and fatty
proteins are admittedly integral proteins of the ER acids. The hydrolysis of TAG by yeasts triacyglycerol
membrane (Yen et al. 2008). However, recent data from lipases provides DAG and FFA for membrane lipid
the oleaginous yeast Rhotorula glutinis indicate that this biosynthesis and the completion of the cell energy needs.
yeast has a soluble homologue of DGA1, reassigned to a The number and function of intracellular lipases differs
third, soluble acyltransferase family, called DGAT3. The between species: S. cerevisiae and Candida parapsilosis
enzyme is found to form a complex with acyl-ACP possess three LB bound lipases (Tgl3p, Tgl4p, and Tgl5p),
synthetase, LPA acyltransferase, and PA phosphatase, each one with different affinity towards TAG, DAG, and
which may be advantageous for the cell in order to produce MAG (Kurat et al. 2006). Y. lipolytica, on the other hand,
TAG near the site of FA neosynthesis (Gangar et al. 2001). may possess only two LB bound triacyglycerol lipases
This finding suggests that a cytosolic pathway of TAG (Tgl3p and Tgl4p), but, being more specialized in HS
synthesis may exist in this oleaginous yeast. In S. degradation, has 16 identified lipase coding genes (encoded
cerevisiae, one DGAT enzyme has been identified, Dga1p, by the LIP genes). Lip2p is secreted in the presence of
belonging to the DGAT2 diacylglycerol:acyltransferase exogenous TAG, while Lip7p and Lip8p are membrane
1200 Appl Microbiol Biotechnol (2011) 90:1193–1206

bound with different substrate specificities (Pignede et al. reaction sequence resulting in a two-carbon shortening
2000; Fickers et al. 2005b). In addition, four more genes (instead of lengthening) of the fatty acid. The cycle can
encoding lipases of the esterase protein family are predicted then be repeated until complete breakdown of the FA to
from genomic data in Y. lipolytica, adding up to 22 lipase acetyl-CoA. However, intermediate fatty acids may leak
proteins. This outnumbers by far the number of lipases from this biochemical route, depending on the physico-
found in other ascomycetous yeasts characterized until now. chemical properties of the specific acyl-CoA and on acetyl-
These enzymes exhibit different substrate specificities and CoA concentrations. This “malfunction” could be exploited
expression patterns, strongly suggesting that they are for aroma production (cyclization of FA) or for polyhy-
regulated depending on TAG content of cells and media droxyalkanoate synthesis after expressing the relevant
(Fickers et al. 2005b; Thevenieau et al. 2009). bacterial enzymes in yeast cells (PHA, see below).
For SE mobilization, not much data are available in The acyl-CoA oxidase gene family carries out the first
yeasts. In S. cerevisiae, three members of the sterol step of β-oxidation, i.e., the formation of a double bond at
hydrolase family (encoded by the YEH1, YLR020/YEH2, the ω-2 position of the acyl-CoA. Gene representation of
and TGL1 genes, respectively) have been identified as this family varies between species. For instance, there are
paralogues to the mammalian acid lipase family (Koffel et three genes in Candida tropicalis, two in Candida maltose,
al. 2005; Mullner et al. 2005). They are localized in the and only one in S. cerevisiae. In Y. lipolytica, six different
plasma and LB membrane as well. The triple knockout acyl-CoA oxidases (encoded by the POX1 to POX6 genes)
mutant has completely lost SE hydrolytic activity but is not with different chain length specificities have been identi-
affected in TAG mobilization, demonstrating tight substrate fied, broadening the adaptation capacity of this species for
specificity of these enzymes (Daum et al. 2007). In Y. HS utilization (Wang et al. 1998, 1999; Luo et al. 2002).
lipolytica, a steryl ester hydrolase, homologous to S. Two of these enzymes, encoded by POX1 and POX6, are
cerevisiae’s Tglp, has been identified in LB (Athenstaedt mostly implicated in dicarboxylic acid (DCA) degradation
et al. 2006). (Thevenieau et al. 2009). Furthermore, the POX gene
Fatty acids, once hydrolyzed from their glycerol or sterol products have been proposed to play a major role in
backbone, are either directed towards phospholipid synthe- peroxisomal division in interaction with the membrane-
sis or to the peroxisome where degradation through β- associated peroxin Pex16p (Guo et al. 2003). Modification
oxidation takes place. The transport mechanism, however, of the POX genotype in Y. lipolytica can lead to specific FA
remains unclear. Available data from S. cerevisiae suggest production and can also result in “slim” or “obese”
that long-chain fatty acids are activated in the cytoplasm by phenotypes (see “Enhancement of lipid synthesis and bio-
the fatty acyl-CoA synthetase Faa1p and Faa4p and oil production—the biotechnological approach” section
transported to the peroxisomes by a complex of two ABC below).
transporters, PXA2 (PAT1) and PXA1 (PAT2) (reviewed by In all yeasts studied so far, the multifunctional enzyme
Hiltunen et al. 2003), while medium-chain FA directly enter encoded by the MFE gene catalyzes both the second and
peroxisomes to be activated by the peroxisomal acyl-CoA third step of β-oxidation. This enzyme possesses two
synthetase Faa2p (Black and DiRusso 2007). Disruption of functional domains exhibiting 2-enoyl-CoA hydratase and
PXA1 or PXA2 results in impaired fatty acid oxidation and 3-hydroxyacyl-CoA dehydrogenase activities, respectively.
in decreased cell growth on medium containing long-chain The unsaturated acyl-CoA is therefore successively
fatty acid as the sole carbon source (Hettema et al. 1996; converted to a 3-hydroxyacyl-CoA and then to a 3-
Swartzman et al. 1996). The substrate of the complex is ketoacyl-CoA, which is finally cleaved by the peroxisomal
suggested to be the membrane-associated fatty acyl-CoAs ketoacyl-CoA thiolase (encoded by the POT1 gene),
and the Pxa proteins flip the polar CoA group across the generating an n−2 acyl-CoA and acetyl-CoA during the
membrane (Hettema et al. 1996). Cells lacking the Pxa1p/ fourth and final step of β-oxidation. However, recent
Pxa2p transporter are capable of growth on medium-chain studies in Y. lipolytica identified a second decane-
fatty acids as laurate (C12:0), suggesting that these can inducible peroxisomal acetoacyl-CoA thiolase (encoded
enter the peroxisome before their CoA activation: misloc- by the PAT1 gene), which is thought to participate in the
alization of the fatty acid activation protein Faa2p to the fourth step of β-oxidation (Yamagami et al. 2001). The
cytosol causes activation of medium-chain fatty acids acetyl-CoA formed by β-oxidation is then redirected to the
outside from the peroxisome, and their oxidation becomes mitochondria towards the glyoxylate and the TCA (Krebs)
entirely dependent on the presence of Pxa1p/Pxa2p cycles, forming a sustainable energy recycling procedure.
(Hettema et al. 1996; Hettema and Tabak 2000). Since acetyl-CoA is unable to cross a lipid bilayer, specific
Once inside the peroxisome, the degradation reactions of shuttles involving acyl-carnitine intermediates and specific
acyl-CoAs are reminiscent of those of fatty acid synthesis translocases have to be used. The routes and export
carried out by FAS. This time, however, it entails a four- pathways differ between S. cerevisiae and for instance
Appl Microbiol Biotechnol (2011) 90:1193–1206 1201

Candida albicans, as S. cerevisiae is atypical among yeasts When non-oleaginous microorganisms are placed in such
in being unable to synthesize carnitine (for a review, see nutriment-limiting conditions, cell proliferation obviously
Strijbis and Distel 2010). It is likely that the situation in ceases, but excess carbon is now directed towards trehalose
most oleaginous yeasts will be similar to that described in and glycogen synthesis (Francois and Parrou 2001;
C. albicans which has a complete carnitine biosynthetic Ratledge and Wynn 2002).
pathway (Strijbis et al. 2009). Yeasts under nutriment limitation undergo three phases
Even though β-oxidation occurs in both peroxisomes of growth: the first is cell proliferation (exponential growth
and mitochondria in mammals and most eukaryotes, in phase), then growth slows down when nutriments become
yeasts only Sporobolomyces roseus has been demonstrated limiting and the cells start to accumulate lipids, and finally
to contain all the mitochondrial enzymes required for cells enter stationary phase where lipid accumulation
mitochondrial β-oxidation. However, some yeast species, continues, but β-oxidation also starts in an effort to
including Y. lipolytica, possess mitochondrial acyl-CoA remobilize the carbon stored, until cells can no longer
dehydrogenase, enoyl-CoA hydratase, and 3-ketoacyl CoA produce essential metabolites and switch off most metabol-
thiolase coding sequences, suggesting that at least some β- ic activity. The duration of each phase depends on the C/N
oxidation could be carried out in the mitochondria (Shen ratio if we take the example of nitrogen limitation. In order
and Burger 2009; Shen et al. 2009). The exact role of this to fine tune the C/N ratio, there are different culture options
hypothetical mitochondrial pathway remains to be assessed. such as batch, fed-batch, and continuous fermentation
The possibility to exploit these findings for enhancing modes. Briefly, in batch mode all minerals and carbon
lipid accumulation and customizing lipid products will be substrates are initially mixed in the reactor, with a high
addressed in the next sections, after a short introduction initial C/N ratio to boost lipid accumulation. No further
on how lipid accumulation is initiated in oleaginous monitoring of the C/N ratio is possible in this mode of
microorganisms. culture. Granger and colleagues (Granger 1992) showed
that accumulation in R. glutinis in batch mode with
different C/N ratio could increase lipid production by a
Setting on lipid accumulation factor of three. In the continuous mode, the C/N ratio stays
constant throughout culture and regulation of substrate
It is now well established that cultivation conditions, concentration leads to the fine tuning of growth rate: lower
nitrogen availability, temperature, pH, metal traces, and growth rates promote more extensive lipid accumulation
mineral concentrations all influence lipid accumulation in (Ykema et al. 1986). Finally, fed-batch mode allows a
oleaginous yeasts (Ratledge and Wynn 2002). These begin precise control of nutrient and substrate flow rates. This
to accumulate lipids when an essential element for cell ensures efficient and reproducible lipid production. This
proliferation becomes limiting in the medium and the last fermentation mode is the most accurate and has been
carbon source is present in excess. Once cell proliferation long used for protein production processes; it is indeed the
becomes limiting, carbon is directly channeled into lipid most reliable approach to use in a biotechnological
synthesis with the resulting buildup of neutral lipids. Then, application (Beopoulos et al. 2009a).
in the presence of an excess of carbon source, depletion of
any essential nutriment can be used for setting on lipid
accumulation, like nitrogen, magnesium, zinc, iron, or Enhancement of lipid synthesis and bio-oil
phosphorus. However, nitrogen limitation is generally used production—the biotechnological approach
(often referred to as a high C/N ratio). This is the easiest
condition to control and it results in the most efficient Organism selection, culture monitoring, and nutriment
accumulation induction factors. The reason of this efficien- limitation may all be used to enhance lipid production or
cy lies in an additional regulation affecting the TCA cycle: to alter lipid composition depending on the carbon source
once nitrogen becomes limiting, the cell in order to produce utilized (Athenstaedt et al. 2006), but for commercializing
ammonium breaks down AMP through AMP desaminase oil derivatives from yeasts it just may not be good enough.
(AMP→IMP+NH4+). AMP is a molecule that happens to Many genetic tools are nowadays available, ranging from
be the co-substrate of isocitrate dehydrogenase in the Krebs conventional in vivo genetics to genetic engineering using
(TCA) cycle. The latter is responsible for the conversion of single or multicopy integration of native or heterologous
citrate to isocitrate and the resulting excess of non- genes.
metabolized citrate is transferred in the cytosol via the The first attempts at ameliorating lipid accumulation
citrate/malate shuttle to be converted to acetyl-CoA by required modification of the expression of key enzymes
ACL. Nitrogen limitation induces therefore the excessive involved in lipid metabolism, so as to redirect carbon
production of the main precursor of fatty acid synthesis. supply towards lipid synthesis. We have already addressed
1202 Appl Microbiol Biotechnol (2011) 90:1193–1206

the case of malic enzyme (ME) in various yeast species in substrate specificity. POX2 deletion blocks the use of long-
order to regenerate the NADPH+ cofactor supply for FA chain fatty acids, whereas deletion of POX3 decreases the
synthesis. The same stands for the overexpression of ACL, degradation of short-chain fatty acids. Mutant strains
increasing the acyl-CoA support towards de novo lipid deleted for POX2, POX3, and POX5 result in a “slim”
synthesis. All these attempts gave impressive results in non- phenotype probably due to a feedback regulation of
oleaginous microorganisms (Ratledge and Wynn 2002), but oxidation. On the other hand, strains overexpressing
failed at touching the limiting steps in Y. lipolytica as stated POX2 result in an obese phenotype (Mlickova et al.
above. Another key enzyme that is currently under 2004a). This kind of manipulation is not possible for non-
investigation is acetyl-CoA carboxylase (ACC), which oleaginous species like S. cerevisiae that encode a single
catalyzes the first and rate-limiting step in de novo lipid acyl-CoA oxidase.
synthesis. ACC1/2 overexpression increases lipid accumu- Another approach, already commercialized by Safisis
lation in non-oleaginous yeasts; however, no data are (Lesaffre, France), makes use of the yeasts β-oxidation
available for oleaginous species to verify if the enzymatic pathway in order to produce aromatic compounds like the
activity of ACC is indeed a limiting step. peach aromatic additive γ-decalactone from ricinoleic acid
Concerning the neutral lipid synthesis pathway, deletion (C18:1-OH). The same is achieved by feeding hydroxy
of the glycerol-3-phosphate dehydrogenase gene (GUT2) in fatty acids to a Y. lipolytica pox3-deleted strain in order to
the oleaginous yeast Y. lipolytica undergoing ex novo lipid abolish the degradation capacity of the C10 fatty acid
accumulation in oleic substrate prevented conversion of lactone precursor (Wache et al. 2003).
G3P to DHAP, which resulted in a 3-fold increase in lipid A different application taking advantage of oxidation
accumulation compared to wild-type strain (Beopoulos et pathways in yeast is the production of dicarboxylic acids
al. 2008). The additional overexpression of the GPD1 gene, (DCA), commonly produced by chemical synthesis for
catalyzing this time the conversion of DHAP to G3P, nylon, resins, and adhesives or for biodiesel production. As
resulted in a 4-fold increase in lipid accumulation (Dulermo stated before, yeasts produce DCA as a result of ω-
T., submitted). These modifications combined with dele- oxidation. The main yeast species modified so far for DCA
tions of the six POX genes encoding acyl-CoA oxidases, production using genetic engineering are Candida cloacae,
abolishing β-oxidation, gave rise to an obese yeast capable Candida tropicalis, and Y. lipolytica. The first industrial
of accumulating more than 80% of its mass as lipids. process was developed by Nippon Mining Company
The aforementioned acyltransferase families carry out (Japan) and afterwards exploited by Cathay Biotechnology
the final acylation step of the TAG formation. The differ- of Shanghai (China) and Cognis (Germany) using mutant
ences in substrate specificity and regulation of these strains of C. tropicalis. In this strain, β-oxidation is
enzymes may be linked to some extent to the differences abolished in order to prevent degradation of the DCA,
in lipid accumulation observed between oleaginous yeasts. and the cytochrome P450 monoxygenase and reductase
The assumption made is that the TAG to FFA ratio systems, involved in the hydroxylation step of ω-oxidation,
regulates total lipid accumulation in cells: boosting up are overexpressed (Picataggio et al. 1992). Furthermore,
TAG production by overexpressing these genes could yield three fatty alcohol oxidases from C. tropicalis (FAO1-3
over-accumulating strains. Overexpression of the DGA2 genes) are overexpressed, resulting in an increased produc-
gene under the control of POX2 constitutive promoter in Y. tivity of DCA (Eschenfeldt et al. 2003). Currently, DCA
lipolytica cultivated in oleic acid containing medium production is attempted in Y. lipolytica, by deleting POX
resulted in a 78% increase of the TAG to FFA ratio with genes, overexpressing NADPH-dependent P450 reductase
a simultaneous increase of 63% in overall lipid accumula- and ALK (P450 cytochrome oxidase) genetic context in
tion, compared to wild-type strain (Beopoulos et al., AEM order to improve substrate specificity and specific chain
submitted). Selective deletion or expression of these length DCA (Iida et al. 2000; Thevenieau 2006).
enzymes may lead to TAGs with specific acyl-chain length: Polyunsaturated fatty acid (PUFA) production is also of
Dupont de Nemours currently tries to exploit these paths for great biotechnological interest as these composites are on
biotechnological applications and has recently deposited a high demand for dietary supplements and fish farm nutri-
patent (U.S. Patent 7465565) claiming heterologous ex- ments. Numerous nutritional recommendations have been
pression in Y. lipolytica of DGAT from various oleaginous made for the inclusion of PUFAs in the diet for the
yeasts to alter the quality and increase the quantity of the oil prevention of coronary heart problems and also for the
produced. improvement of retinal and brain functions. Suitable
Modulation of the degradation pathways (ω- and β- production hosts that have been considered are filamentous
oxidation) is also of interest for potential biotechnological fungi like M. circinelloides for γ-linolenic acid (18:3, n−6),
applications. Acyl-CoA oxidase (POX genes) Y. lipolytica Mortierella alpina for arachidonic acid (20:4, n−6), and
mutants display altered lipid profiles as a result of the Aox Crypthecodinium cohnii and Schizochytrium for docosa-
Appl Microbiol Biotechnol (2011) 90:1193–1206 1203

hexaenoic acid (22:6, n−3) production. All these organisms Mammals, however, cannot synthesize carotenoids and
are used, or have been used, commercially to produce these must obtain them through their diet. In general, the natural
SCOs, in stirred tank fermenters of up to 220 m3 capacity biological systems that produce carotenoids are industrially
(Ratledge 2001; Meng et al. 2009). Currently, non- intractable and produce the compounds at such low levels
microbial sources of these materials are favored, includ- that production on a commercial scale is not practicable.
ing many endangered species of fish. But new alter- Therefore, most carotenoids used in industry are produced
natives might be proposed, as shown for instance by by chemical synthesis, generating the need for more
Dupont de Nemours who genetically engineered strains environmental friendly biological processes for their pro-
of Y. lipolytica (patent WO/2006/052871) in order to duction. Some efforts have previously been made to
commercialize docosahexaenoic acid (DHA) as an alter- genetically engineer bacteria or fungi to produce high
native source to fish oil. Although Y. lipolytica accumu- levels of carotenoids (Visser et al. 2003). Biosynthesis of
lates lipids to lower levels than other oleaginous species, it these pigments in non-carotenogenic hosts such as yeasts
is the only yeast known to accumulate a high proportion of requires the heterologous expression of a geranylgeranyl
linoleic acid (more than 50% of the total FA—Table 1), diphosphate synthetase encoding gene (crtE) to comple-
which is the intermediate substrate of DHA synthesis. The ment their own isoprenoid pathway. This enzyme catalyses
DHA-producing strain of Y. lipolytica expresses Δ4, Δ5, the head-to-tail condensation of an isoprenoid unit to the
Δ6, and Δ7 desaturases of the oleaginous fungus M. isoprenoid precursor farnesyl diphosphate (C15), yielding
alpina, the C18–20, C20–22 elongases of the heterotro- geranylgeranyl diphosphate (C20, GGDP). An additional
phic marine protist Thraustochytrium aureum and is recombinant enzyme, phytoene synthetase (CrtB), is then
genetically modified for its TAG acyltransferases. Recent- required for the head-to-head condensation of two GGDP
ly, a Dupont de Nemours Y. lipolytica strain producing up molecules to the colorless C40 carotenoid phytoene
to 34% (w/w) of eicosapentaenoic acid (EPA) was (Sabirova et al. 2010). Expression of these enzymes in the
characterized as safe, matching the safety profile of the host Escherichia coli allowed the recombinant biosynthesis
EPA commercial fish oil (Belcher et al. 2011). Other of diverse cyclic and acyclic carotenoids as well (Schmidt-
studies target the production of ω-3 and ω-6 PUFA as γ- Dannert 2000). In the need of improved systems, allowing
linolenic acid (Damude et al. 2006). higher levels of production and facilitated purification,
Polyhydroxyalkanoates (PHA) are polyesters (bioplas- efforts have been devoted to carotenoid production in
tics) with interesting thermoplastic and elastomeric proper- oleaginous yeasts such as Y. lipolytica (Ailey et al. 2007).
ties. The 3-hydroxyacyl-CoA intermediate of β-oxidation An interesting review presenting novel ideas for biocon-
(see above) is the substrate of PHA synthetase, a protein version of fats and lipids by expressing bacterial genes in Y.
found in a number of bacteria, but absent from yeasts. The lipolytica was recently published (Sabirova et al. 2010).
synthesis of medium chain length (MCL) PHA has been
achieved through peroxisomal targeting of the PHA
synthetase from Pseudomonas aeruginosa in S. cerevisiae Future works and perspectives
and Pichia pastoris (Poirier et al. 2001, 2002). The size of
the PHA monomers synthesized by these yeasts ranges Negative environmental impact of fossil fuels and the need
from five to 14 carbons and is generated by the degradation of renewable energy have prompted research towards
of odd- or even-chain fatty acids via the peroxisomal β- biofuel production. The high cost and price imbalance
oxidation cycle. Recently, Haddouche and colleagues created by using plant oils or animal fat as petroleum
expressed the PHA synthetase from P. aeruginosa in substitutes aroused interest in developing oleaginous micro-
different POX genetic backgrounds of Y. lipolytica, result- organisms to obtain large amounts of oils for biodiesel
ing in high yields of PHA production, depending on the utilization. However, producing competitive biodiesel is
POX genotype (Haddouche et al. 2010). Further improve- possible only if the oil is produced from cheap raw
ments of PHA production by yeasts will require the materials as substrates. In the future, biofuel production
modification of the MFE enzyme in order to block β- may combine use of plant leftovers to oil production by
oxidation after the production of 3-hydroxyacyl-CoA, the yeasts. It would entail the screening for oleaginous yeast
immediate substrate of PHA synthetase. strains tolerant to lignocellulose degradation products
Finally, carotenoid production has been considered in (Chen et al. 2009) and evolutionary engineering of yeasts.
yeasts. Carotenoids represent a large group of structurally Such an approach has already been developed in S.
diverse pigments, with an acyl chain ranging from C30 to cerevisiae to improve xylose fermentation for ethanol
C50 carbons, synthesized by microorganisms and plants. production (Garcia Sanchez et al. 2010).
They are commercially used as food colorants, nutraceut- Further optimization of yeast potentials for oil produc-
icals, and for cosmetic and pharmaceutical purposes. tion by means of substrate selection and genetic or
1204 Appl Microbiol Biotechnol (2011) 90:1193–1206

metabolic engineering is crucial for the development of Biobel F, Erdmann R (1996) Identification of a yeast peroxisomal
member of the family of AMP-binding proteins. Eur J Biochem
future biotechnological applications. It remains essential
340:468–476
that the metabolism of more yeasts be studied as new Black PN, DiRusso CC (2007) Vectorial acylation: linking fatty acid
sequence data and more genetic tools become available. transport and activation to metabolic trafficking. Novartis Found
This will lead to the understanding and the exploitation of Symp 286:127–138, discussion 138–141, 162–123, 196–203
Boer E, Steinborn G, Kunze G, Gellissen G (2007) Yeast expression
the mechanisms conferring massive lipid accumulation and
platforms. Appl Microbiol Biotechnol 77:513–523
specific oil production in certain species. New methods Boles E, de Jong-Gubbels P, Pronk JT (1998) Identification and
combining lipidomic, metabolomic, and genetic approaches characterization of mae1, the Saccharomyces cerevisiae structural
will undoubtedly provide much information about the gene encoding mitochondrial malic enzyme. J Bacteriol
180:2875–2882
regulation of lipid metabolism. Combining these informa-
Botham PA, Ratledge C (1979) A biochemical explanation for lipid
tions with subcellular localization data and feeding them accumulation in Candida 107 and other oleaginous micro-
into models of the lipid pathways and their regulation may organisms. J Gen Microbiol 114:361–375
provide novel ideas (Boer et al. 2007). In addition, making Brown DA (2001) Lipid droplets: proteins floating on a pool of fat.
Curr Biol 11:446–449
use of the byproducts generated in the course of fatty acid
Chen X, Li Z, Zhang X, Hu F, Ryu DD, Bao J (2009) Screening of
production to manufacture proteins, nutrients, steroids, and oleaginous yeast strains tolerant to lignocellulose degradation
aromatic components may reduce the production cost of the compounds. Appl Biochem Biotechnol 159:591–604
lipid moieties and constitute a side biotechnological Chirala SS (1992) Coordinated regulation and inositol-mediated and fatty
acid-mediated repression of fatty acid synthase genes in Saccharo-
application on its own.
myces cerevisiae. Proc Natl Acad Sci USA 89:10232–10236
In conclusion, even though few detailed studies in yeast Coleman RA, Lee DP (2004) Enzymes of triacylglycerol synthesis
species other than S. cerevisiae and Y. lipolytica have been and their regulation. Prog Lipid Res 43:134–176
carried out, increasing efforts are devoted towards the Czabany T, Athenstaedt K, Daum G (2007) Synthesis, storage and
manipulation and regulation of yeast lipid metabolism for degradation of neutral lipids in yeasts. Biochim Biophys Acta
1771:299–309
bio-oil production, building up an emergent biotechnolog- Dahlqvist A, Stahl U, Lenman M, Banas A, Lee M, Sandager L,
ical field with many promising applications. Ronne H, Stymne S (2000) Phospholipid:diacylglycerol acyl-
transferase: an enzyme that catalyzes the acyl-CoA-independent
formation of triacylglycerol in yeast and plants. Proc Natl Acad
References Sci USA 97:6487–6492
Damude HG, Zhang H, Farrall L, Ripp KG, Tomb JF, Hollerbach D,
Yadav NS (2006) Identification of bifunctional delta12/omega3
Ailey R, Madden K, Trueheart J (2007) Production of carotenoids in fatty acid desaturases for improving the ratio of omega3 to
oleaginous yeast and fungi. United States Patent No omega6 fatty acids in microbes and plants. Proc Natl Acad Sci
20070015237 USA 103:9446–9451
Amaretti A, Raimondi S, Sala M, Roncaglia L, De Lucia M, Leonardi Daum G, Wagner A, Czabany T, Athenstaedt K (2007) Dynamics of
A, Rossi M (2010) Single cell oils from the cold adapted neutral lipid storage and mobilization in the yeast. Biochimie
oleaginous yeast Rhotorula glacialis DBVPG 4785. Micr Cell 89:243–248
Fact 9:73 Dell'Angelica EC, Stella CA, Ermacora MR, Ramos EH, Santome JA
Athenstaedt K, Daum G (2006) The life cycle of neutral lipids: (1992) Study on fatty acid binding by proteins in yeast.
synthesis, storage and degradation. Cell Mol Life Sci 63:1355– Dissimilar results in Saccharomyces cerevisiae and Yarrowia
1369 lipolytica. Comp Biochem Physiol B 102:261–265
Athenstaedt K, Jolivet P, Boulard C, Zivy M, Negroni L, Nicaud JM, Denic V, Weissman JS (2007) A molecular caliper mechanism for
Chardot T (2006) Lipid particle composition of the yeast determining very long-chain fatty acid length. Cell 130:663–677
Yarrowia lipolytica depends on the carbon source. Proteomics 6 DiRusso CC, Black PN (1999) Long-chain fatty acid transport in
(5):1450–1459 bacteria and yeast. Paradigms for defining the mechanism
Beckman M (2006) Cell biology. Great balls of fat. Science 311 underlying this protein-mediated process. Mol Cell Biochem
(5765):1232–1234 192(1–2):41–52
Belcher LA, Mackenzie SA, Donner M, Sykes GP, Frame SR, Gillies Dujon B, Sherman D, Fischer G, Durrens P, Casaregola S, Lafontaine
PJ (2011) Safety assessment of EPA-rich triglyceride oil I, de Montigny J, Marck C, Neuveglise C, Talla E, Goffard N,
produced from yeast: genotoxicity and 28-day oral toxicity in Frangeul L, Aigle M, Anthouard V, Babour A, Barbe V, Barnay
rats. Regul Toxicol Pharmacol 59:53–63 S, Blanchin S, Beckerich J-M, Beyne E, Bleykasten C, Boisrame
Beopoulos A, Mrozova Z, Thevenieau F, Le Dall MT, Hapala I, A, Boyer J, Cattolico L, Confanioleri F, de Daruvar A, Despons
Papanikolaou S, Chardot T, Nicaud JM (2008) Control of lipid L, Fabre E, Fairhead C, Ferry-Dumazet H, Groppi A, Hantraye F,
accumulation in the yeast Yarrowia lipolytica. Appl Environ Hennequin C, Jauniaux N, Joyet P, Kachouri R, Kerrest A,
Microbiol 74:7779–7789 Koszul R, Lemaire M, Lesur I, Ma L, Muller H, Nicaud J-M,
Beopoulos A, Cescut J, Haddouche R, Uribelarrea JL, Molina-Jouve Nikolski M, Oztas S, Ozier-Kalogeropoulos O, Pellenz S, Potier
C, Nicaud JM (2009a) Yarrowia lipolytica as a model for bio-oil S, Richard G-F, Straub M-L, Suleau A, Swennen D, Tekaia F,
production. Prog Lipid Res 48:375–387 Wesolowski-Louvel M, Westhof E, Wirth B, Zeniou-Meyer M,
Beopoulos A, Desfougeres T, Sabirova J, Zinjarde S, Neuveglise C, Zivanovic I, Bolotin-Fukuhara M, Thierry A, Bouchier C,
Nicaud JM (2009b) The hydrocarbon-degrading oleaginous yeast Caudron B, Scarpelli C, Gaillardin C, Weissenbach J, Wincker
Yarrowia lipolytica. In: Timmis KN (ed) Handbook of hydrocar- P, Souciet J-L (2004) Genome evolution in yeasts. Nature 430
bon and lipid microbiology. Springer, Germany (6995):35–44
Appl Microbiol Biotechnol (2011) 90:1193–1206 1205

Eschenfeldt WH, Zhang Y, Samaha H, Stols L, Eirich LD, Wilson CR, Hettema EH, Tabak HF (2000) Transport of fatty acids and
Donnelly MI (2003) Transformation of fatty acids catalyzed by metabolites across the peroxisomal membrane. Biochim Biophys
cytochrome p450 monooxygenase enzymes of Candida tropica- Acta 1486:18–27
lis. Appl Environ Microbiol 69:5992–5999 Hettema EH, van Roermund CW, Distel B, van den Berg M, Vilela C,
Faergeman NJ, DiRusso CC, Elberger A, Knudsen J, Black PN (1997) Rodrigues-Pousada C, Wanders RJ, Tabak HF (1996) The abc
Disruption of the Saccharomyces cerevisiae homologue to the transporter proteins pat1 and pat2 are required for import of long-
murine fatty acid transport protein impairs uptake and growth on chain fatty acids into peroxisomes of Saccharomyces cerevisiae.
long-chain fatty acids. J Biol Chem 272:8531–8538 EMBO J 15:3813–3822
Ferreyra RG, Burgardt NI, Milikowski D, Melen G, Kornblihtt AR, Hiltunen JK, Mursula AM, Rottensteiner H, Wierenga RK, Kastaniotis
Dell' Angelica EC, Santome JA, Ermacora MR (2006) A yeast AJ, Gurvitz A (2003) The biochemistry of peroxisomal [beta]-
sterol carrier protein with fatty-acid and fatty-acyl-CoA binding oxidation in the yeast Saccharomyces cerevisiae. FEMS Microbiol
activity. Arch Biochem Biophys 453:197–206 Rev 27:35–64
Fickers P, Benetti PH, Wache Y, Marty A, Mauersberger S, Smit MS, Hiltunen JK, Schonauer MS, Autio KJ, Mittelmeier TM, Kastaniotis
Nicaud JM (2005a) Hydrophobic substrate utilisation by the AJ, Dieckmann CL (2009) Mitochondrial fatty acid synthesis
yeast Yarrowia lipolytica, and its potential applications. FEMS type ii: more than just fatty acids. J Biol Chem 284:9011–9015
Yeast Res 5(6–7):527–543 Holdsworth JE, Veenhuis M, Ratledge C (1988) Enzyme activities in
Fickers P, Fudalej F, Le Dall MT, Casaregola S, Gaillardin C, oleaginous yeasts accumulating and utilizing exogenous or
Thonart P, Nicaud JM (2005b) Identification and character- endogenous lipids. J Gen Microbiol 134:2907–2915
isation of lip7 and lip8 genes encoding two extracellular Iida T, Sumita T, Ohta A, Takagi M (2000) The cytochrome p450alk
triacylglycerol lipases in the yeast Yarrowia lipolytica. Fungal multigene family of an n-alkane-assimilating yeast, Yarrowia
Genet Biol 42:264–274 lipolytica: cloning and characterization of genes coding for new
Francois J, Parrou JL (2001) Reserve carbohydrates metabolism in the cyp52 family members. Yeast 16:1077–1087
yeast Saccharomyces cerevisiae. FEMS Microbiol Rev 25:125–145 Jacquier N, Schneiter R (2010) Ypk1, the yeast orthologue of the
Fujimoto T, Ohsaki Y, Cheng J, Suzuki M, Shinohara Y (2008) Lipid human serum- and glucocorticoid-induced kinase, is required for
droplets: a classic organelle with new outfits. Histochem Cell efficient uptake of fatty acids. J Cell Sci 123:2218–2227
Biol 130:263–279 Kihara A, Sakuraba H, Ikeda M, Denpoh A, Igarashi Y (2008)
Gangar A, Karande AA, Rajasekharan R (2001) Isolation and Membrane topology and essential amino acid residues of Phs1, a
localization of a cytosolic 10 s triacylglycerol biosynthetic 3-hydroxyl-CoA dehydratase involved in very long-chain fatty
multienzyme complex from oleaginous yeast. J Biol Chem acid elongation. J Biol Chem 283:11199–11209
276:10290–10298 Koffel R, Tiwari R, Falquet L, Schneiter R (2005) The Saccharomyces
Garcia Sanchez R, Karhumaa K, Fonseca C, Sanchez Nogue V, cerevisiae YLL012/YEH1, YLR020/YEH2, and TGL1 genes
Almeida JR, Larsson CU, Bengtsson O, Bettiga M, Hahn- encode a novel family of membrane-anchored lipases that are
Hagerdal B, Gorwa-Grauslund MF (2010) Improved xylose and required for steryl ester hydrolysis. Mol Cell Biol 25:1655–1668
arabinose utilization by an industrial recombinant Saccharomyces Kohlwein SD (2010) Triacylglycerol homeostasis: insights from yeast.
cerevisiae strain using evolutionary engineering. Biotechnol J Biol Chem 285(21):15663–15667
Biofuels 3:13 Kohlwein SD, Eder S, Oh CS, Martin CE, Gable K, Bacikova D,
Gaspar ML, Hofbauer HF, Kohlwein SD, Henry SA (2011) Coordi- Dunn T (2001) Tsc13p is required for fatty acid elongation
nation of storage lipid synthesis and membrane biogenesis: and localizes to a novel structure at the nuclear-vacuolar
evidence for cross-talk between triacylglycerol metabolism and interface in Saccharomyces cerevisiae. Mol Cell Biol 21:109–
phosphatidylinositol synthesis. J Biol Chem 286:1696–1708 125
Goodrich-Tanrikulu M, Stafford AE, Lin JT, Makapugay MI, Fuller Kurat CF, Natter K, Petschnigg J, Wolinski H, Scheuringer K, Scholz
G, McKeon TA (1994) Fatty acid biosynthesis in novel ufa H, Zimmermann R, Leber R, Zechner R, Kohlwein SD (2006)
mutants of Neurospora crassa. Microbiol 140(10):2683–2690 Obese yeast: triglyceride lipolysis is functionally conserved from
Granger L (1992) Caractérisation cinétique et stoechiometrique de la mammals to yeast. J Biol Chem 281:491–500
synthèse d’acide gras chez Rhodotorula glutinis. Institut National Luo YS, Nicaud JM, Van Veldhoven PP, Chardot T (2002) The acyl-
des sciences appliquées de Toulouse, Toulouse CoA oxidases from the yeast Yarrowia lipolytica: characteriza-
Gui M, Lee KT, Bhatia S (2008) Feasibility of edible oil vs. non- tion of aox2p. Arch Biochem Biophys 407:32–38
edible oil vs. waste edible oil as biodiesel feedstock. Energy Meng X, Jianming Y, Xin X, Lei Z, Qingjuan N, Mo X (2009)
33:1646–1653 Biodiesel production from oleaginous microorganisms. Renew-
Guo T, Kit YY, Nicaud JM, Le Dall MT, Sears SK, Vali H, Chan H, able Energy 34:1–5
Rachubinski RA, Titorenko VI (2003) Peroxisome division in the Mlickova K, Luo Y, D'Andrea S, Pec P, Chardot T, Nicaud J (2004a)
yeast Yarrowia lipolytica is regulated by a signal from inside the Acyl-CoA oxidase, a key step for lipid accumulation in the yeast
peroxisome. J Cell Biol 162:1255–1266 Yarrowia lipolytica. J Mol Catal B Enzym 28:81–85
Haddouche R, Delessert S, Sabirova J, Neuveglise C, Poirier Y, Nicaud Mlickova K, Roux E, Athenstaedt K, d'Andrea S, Daum G, Chardot T,
JM (2010) Roles of multiple acyl-CoA oxidases in the routing of Nicaud JM (2004b) Lipid accumulation, lipid body formation,
carbon flow towards beta-oxidation and polyhydroxyalkanoate and acyl coenzyme a oxidases of the yeast Yarrowia lipolytica.
biosynthesis in Yarrowia lipolytica. FEMS Yeast Res 10:917–927 Appl Environ Microbiol 70:3918–3924
Hasslacher M, Ivessa AS, Paltauf F, Kohlwein SD (1993) Acetyl-CoA Mullner H, Deutsh G, Leitner E, Ingolic E (2005) YEH2/YLR020c
carboxylase from yeast is an essential enzyme and is regulated by encodes a novel steryl ester hydrolase of the yeast Saccharomy-
factors that control phospholipid metabolism. J Biol Chem ces cerevisiae. J Biol Chem 280:13321–13328
268:10946–10952 Nielsen J (2009) Systems biology of lipid metabolism: from yeast to
Heier C, Taschler U, Rengachari S, Oberer M, Wolinski H, Natter K, human. FEBS Lett 583:3905–3913
Kohlwein SD, Leber R, Zimmermann R (2010) Identification of Picataggio S, Rohrer T, Deanda K, Lanning D, Reynolds R, Mielenz J,
yju3p as functional orthologue of mammalian monoglyceride Eirich LD (1992) Metabolic engineering of Candida tropicalis
lipase in the yeast Saccharomyces cerevisiae. Biochim Biophys for the production of long-chain dicarboxylic acids. Nat
Acta 1801:1063–1071 Biotechnol (N Y) 10:894–898
1206 Appl Microbiol Biotechnol (2011) 90:1193–1206

Pignede G, Wang H, Fudalej F, Gaillardin C, Seman M, Nicaud JM Swartzman EE, Viswanathan MN, Thorner J (1996) The pal1 gene
(2000) Characterization of an extracellular lipase encoded by lip2 product is a peroxisomal atp-binding cassette transporter in the
in Yarrowia lipolytica. J Bacteriol 182:2802–2810 yeast Saccharomyces cerevisiae. J Cell Biol 132:549–563
Poirier Y, Erard N, Petetot JM (2001) Synthesis of polyhydroxyalka- Tang W, Zhang S, Tan H, Zhao ZK (2010) Molecular cloning and
noate in the peroxisome of Saccharomyces cerevisiae by using characterization of a malic enzyme gene from the oleaginous
intermediates of fatty acid beta-oxidation. Appl Environ Micro- yeast Lipomyces starkeyi. Mol Biotechnol 45:121–128
biol 67:5254–5260 Tehlivets O, Scheuringer K, Kohlwein SD (2007) Fatty acid synthesis
Poirier Y, Erard N, MacDonald-Comber PJ (2002) Synthesis of and elongation in yeast. Biochim Biophys Acta 1771:255–270
polyhydroxyalkanoate in the peroxisome of Pichia pastoris. Theodoulou FL, Holdsworth M, Baker A (2006) Peroxisomal abc
FEMS Microbiol Lett 207:97–102 transporters. FEBS Lett 580:1139–1155
Rajakumari S, Grillitsch K, Daum G (2008) Synthesis and turnover of Thevenieau F (2006) Metabolic engineering of the yeast Yarrowia
non-polar lipids in yeast. Prog Lipid Res 47:157–171 lipolytica for the production of long-chain dicarboxylic acids
Ratledge C (1989) Microbial lipids, vol 2. Academic, London from renewable oil feedstock. Ph.D. thesis
Ratledge C (1994) Yeasts, moulds, algae and bacteria as sources of Thevenieau F, Le Dall MT, Nthangeni B, Mauersberger S, Marchal R,
lipids. In: Kamel BS, Kakuda Y (eds) Technological advances in Nicaud JM (2007) Characterization of Yarrowia lipolytica
improved and alternative sources of lipids. Blackie Academic and mutants affected in hydrophobic substrate utilization. Fungal
Professional, London, pp 235–291 Genet Biol 44:531–542
Ratledge C (2001) Structure and modified lipids. Marcel Dekker, New Thevenieau F, Beopoulos A, Desfougeres T, Sabirova J, Albertin K,
York Zinjarde S, Nicaud JM (2009) Uptake and assimilation of
Ratledge C (2004) Fatty acid biosynthesis in microorganisms being hydrophobic substrates by the oleaginous yeast Yarrowia lip-
used for single cell oil production. Biochimie 86:807–815 olytica. In: Timmis KN (ed) Handbook of hydrocarbon and lipid
Ratledge C, Wynn JP (2002) The biochemistry and molecular biology microbiology. Springer, Berlin
of lipid accumulation in oleaginous microorganisms. Adv Appl Thurmond W (2008) Biodiesel 2020: a global market survey.
Microbiol 51:1–51 Emerging markets
Ratlege C, Tan K (1990) Oils and fats: production, degradation and Visser H, van Ooyen AJ, Verdoes JC (2003) Metabolic engineering of
utilization by yeasts. In: Verachtert H, De Mot R (eds) Yeast the astaxanthin-biosynthetic pathway of Xanthophyllomyces
biotechnology and biocatalysis. Marcel Dekker, New York, pp dendrorhous. FEMS Yeast Res 4:221–231
223–254 Wache Y, Aguedo M, Nicaud JM, Belin JM (2003) Catabolism of
Rattray JBM (1988) Microbial lipids, vol 1. Academic, London hydroxyacids and biotechnological production of lactones by
Ruenwai R, Cheevadhanarak S, Laoteng K (2009) Overexpression of Yarrowia lipolytica. Appl Microbiol Biotechnol 61:393–404
acetyl-CoA carboxylase gene of Mucor rouxii enhanced fatty acid Wang H, Le Clainche A, Le Dall MT, Wache Y, Pagot Y, Belin JM,
content in Hansenula polymorpha. Mol Biotechnol 42:327–332 Gaillardin C, Nicaud JM (1998) Cloning and characterization of
Sabirova J, Haddouche R, Van Bogaert I, Mulaa F, Verstraete W, the peroxisomal acyl CoA oxidase aco3 gene from the alkane-
Timmis K, Schmidt-Dannert C, Nicaud JM, Soetaert W (2010) utilizing yeast Yarrowia lipolytica. Yeast 14:1373–1386
The lipoyeasts project: using the oleaginous yeast Y. lipolytica in Wang H, Le Dall MT, Wache Y, Laroche C, Belin JM, Nicaud JM
combination with specific bacterial genes for the bioconversion (1999) Cloning, sequencing, and characterization of five genes
of lipids, fats and oils into high value products. Microb coding for acyl-CoA oxidase isozymes in the yeast Yarrowia
Biotechnol 4:47–54 lipolytica. Cell Biochem Biophys 31:165–174
Sandager L, Gustavsson MH, Stahl U, Dahlqvist A, Wiberg E, Banas Wynn JP, Kendrick A, Ratledge C (1997) Sesamol as an inhibitor of
A, Lenman M, Ronne H, Stymne S (2002) Storage lipid growth and lipid metabolism in Mucor circinelloides via its
synthesis is non-essential in yeast. J Biol Chem 277:6478–6482 action on malic enzyme. Lipids 32:605–610
Schmidt-Dannert C (2000) Engineering novel carotenoids in micro- Yamagami S, Iida T, Nagata Y, Ohta A, Takagi M (2001) Isolation and
organisms. Curr Opin Biotechnol 11:255–261 characterization of acetoacetyl-CoA thiolase gene essential for n-
Schweizer E, Hofmann J (2004) Microbial type i fatty acid synthases decane assimilation in yeast Yarrowia lipolytica. Biochem
(fas): major players in a network of cellular fas systems. Biophys Res Commun 28:832–838
Microbiol Mol Biol Rev 68:501–517 Yen CL, Stone SJ, Koliwad S, Harris C, Farese RV Jr (2008) Thematic
Shen YQ, Burger G (2009) Plasticity of a key metabolic pathway in review series: glycerolipids. Dgat enzymes and triacylglycerol
fungi. Funct Integr Genomics 9:145–151 biosynthesis. J Lipid Res 49:2283–2301
Shen YQ, Lang BF, Burger G (2009) Diversity and dispersal of a Ykema A, Verbree EC, van Verseveld HW, Smit H (1986)
ubiquitous protein family: acyl-CoA dehydrogenases. Nucleic Mathematical modelling of lipid production by oleaginous yeasts
Acids Res 37:5619–5631 in continuous cultures. Antonie Leeuwenhoek 52:491–506
Steen EJ, Kang Y, Bokinsky G, Hu Z, Schirmer A, McClure A, Del Zhang Y, Adams IP, Ratledge C (2007) Malic enzyme: the controlling
Cardayre SB, Keasling JD (2010) Microbial production of fatty-acid- activity for lipid production? Overexpression of malic enzyme in
derived fuels and chemicals from plant biomass. Nature 463:559–562 Mucor circinelloides leads to a 2.5-fold increase in lipid
Strijbis K, Distel B (2010) Intracellular acetyl unit transport in fungal accumulation. Microbiology 153:2013–2025
carbon metabolism. Eukaryot Cell 9:1809–1815 Zou Z, DiRusso CC, Ctrnacta V, Black PN (2002) Fatty acid transport
Strijbis K, van Roermund CW, Hardy GP, van den Burg J, Bloem K, in Saccharomyces cerevisiae. Directed mutagenesis of FAT1
de Haan J, van Vlies N, Wanders RJ, Vaz FM, Distel B (2009) distinguishes the biochemical activities associated with Fat1p. J
Identification and characterization of a complete carnitine Biol Chem 277:31062–31071
biosynthesis pathway in Candida albicans. FASEB J 23:2349– Zweytick D, Athenstaedt K, Daum G (2000) Intracellular lipid particles
2359 of eukaryotic cells. Biochim Biophys Acta 1469:101–120

You might also like