You are on page 1of 17

Biotechnology Advances 48 (2021) 107725

Contents lists available at ScienceDirect

Biotechnology Advances

journal homepage: www.elsevier.com/locate/biotechadv

Research review paper

Biotechnological production of lipid and terpenoid from thraustochytrids


Fei Du a,1, Yu-Zhou Wang a,1, Ying-Shuang Xu a, Tian-Qiong Shi a, Wen-Zheng Liu a, Xiao-Man Sun a,*, He
Huang a,b
a
School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, People’s Republic of China b College
of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing, People’s Republic of China

ARTICLEINFO ABSTRACT
Keywords: As fungus-like protists, thraustochytrids have been increasingly studied for their faster growth rates and high lipid content. In
Thraustochytrids the 1990s, thraustochytrids were used as docosahexaenoic acid (DHA) producers for the first time. Thraustochytrids genera,
Lipid such as Thraustochytrium, Schizochytrium, and Aurantiochytrium have been developed and patented as industrial strains for
Metabolic engineering DHA production. The high DHA yield is attributed to its unique and efficient polyketide-like synthase (PKS) pathway.
Downstream processing Moreover, thraustochytrids possess a completed mevalonate (MVA) pathway, so it can be used as host for terpenoid
production. In order to improve strain performance, the metabolic engineering strategies have been applied to promote or
disrupt intracellular metabolic pathways, such as genetic engineering and addition of chemical activators. However, it is
difficult to realize industrialization only by improving strain performance. Various operation strategies were developed to
enlarge the production quantities from the laboratory-scale, including two-stage cultivation strategies, scale-up technologies
and bioreactor design. Moreover, an economical and effective downstream process is also an important consideration for the
industrial application of thraustochytrids. Downstream costs accounts for 20–60% of the overall process costs, which
represents an attractive target for increasing the cost-competitiveness of thraustochytrids, including how to improve the
efficiency of lipid extraction and the further application of biomass residues. This review aims to overview the whole lipid
biotechnology of thraustochytrids to provide the background information for researchers.

1. Introduction Li et al., 2015). The main fatty acids of most thraustochytrids


include C14:0, C16:0, docosapentaenoic acid (DPA) and DHA, and
Thraustochytrids are marine unicellular heterotrophic the total content of C16:0 and DHA occupy more than 65% of
microorganisms. Previously, thraustochytrids were classified as total fatty acids (Jiang et al., 2004; Meesapyodsuk and Qiu, 2016).
microalgae, but taxonomists rarely used this classification Therefore, the lipid profiles was introduced into the classification
because they have no plastids and are incapable of of thraustochytrids as biochemical characteristics. Based on this,
photosynthesis (Leyland et al., 2017). Nine genera of thraustochytrids can be separated into 5 major groups, including
thraustochytrids have been recognized to date, including DHA/docosapentaenoic acid (DPA), DHA/DPA/eicosapentaenoic
Thraustochytrium, Schizochytrium (Goldstein and Belsky, 1964), acid (EPA), DHA/EPA, DHA/DPA/ EPA/arachidonic acid (ARA), and
Aurantiochytrium (Yokoyama and Honda, 2007), Japonochytrium, DHA/DPA/EPA/ARA/docosate-
Ulkenia, Monorhizochytrium (Doi and Honda, 2017), traenoic acid. And this new classification is consistent with the

* Corresponding author at: School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, People’s Republic of China.
E-mail address: xiaomansun@njnu.edu.cn (X.-M. Sun).
1
These two authors contributed equally to this work. https://doi.org/10.1016/j.biotechadv.2021.107725

Received 3 November 2020; Received in revised form 15 January 2021; Accepted 25 February 2021 Available online 13
March 2021
0734-9750/© 2021 Elsevier Inc. All rights reserved.

Sicyoidochytrium, Parietichytrium, and Botryochytrium result of 18S rRNA gene classification, which provide great
(Yokoyama and Honda, 2007). Thraustochytrids attracted models for studying the mechanism PUFA biosynthesis by the
biotechnological interest because the lipid content of some genetic approach (Huang et al., 2003).
thraustochytrid strains can reach more than 55% of dry cell DHA has attracted great attention as essential for the
weight (DCW) under optimized conditions (Jakobsen et al., 2008; development of eyes and neuron of infants (Innis, 2008), and the
F. Du et al. Biotechnology Advances 48 (2021) 107725
infant formulae is the fastest-growing segment for DHA fermentation conditions, so thraustochytrids are highly
consumption, with a 14% of growth interesting chassis cells for further systematic metabolic
rate from 2015 to 2025 (Ana et al., 2016). DHA is traditionally engineering.
obtained from fish oil, but mass fishing has become There are many other oleaginous microorganisms with an
unsustainable and unable to meet the demand for DHA in recent inherent capacity to produce excess amounts of fatty acids and
years, suggesting the need to find new sustainable resources. store them inside the cell in the form of triacylglycerols (TAGs).
Currently, the largest commercial production of microbial DHA is By engineering the intracellular redox metabolism, the highest
produced by Schizochytrium species, due to it has been approved reported lipid titer of Yarrowia lipolytica reached to 98.9 g/L
by FDA (Ratledge, 2012). DSM is the market leader for microbial (Qiao et al., 2017). Moreover, oleaginous microalgae have been
DHA production from thraustochytrids, including DHAGold and frequently employed for photosynthetic lipid production from
Life’sDHA. In 2018, DSM and Evonik industries established a joint CO2, but a major drawback of this approach is the low
venture to produce DHA and EPA from Schizochytrium, aimed to photosynthetic grow rate of the commonly used organisms such
meet about 15% of the global farmed salmon demand for as Chlorella vulgaris and Synechocystis sp. (Chu et al., 2020, Liang
omega-3 algal oil (https://cen.acs.org/articles/95/i11/DSM- and Lindblad, 2017). By contrast, thraustochytrids can produce
Evonik-form-omega-3-join t-venture.html). Moreover, many high biomass yields of 165 g/L, along with lipid titers of 113 g/L
thraustochytrids species with higher saturated fatty acids (SFA)- without any genetic modification (Guo et al., 2017). In this
producing capacity were isolated. The 54% and 44% content of review, technologies applied in thraustochytrids to produce lipid
SFAs was found in thraustochytrids that screened from the and terpenoid are summarized and discussed, including low-cost
environment in Australia and UK, respectively (Lee Chang et al., cultivation, molecular approaches, engineering operation
2013; Marchan et al., 2017). Recent, thraustochytrid strains with strategies and downstream processing technologies (Fig. 1).
high SFA production were isolated from the coastal waters of
Shenzhen, China, with SFA content of 62% (Wang et al., 2018b). 2. The biosynthetic pathways of lipid and terpenoid
The quality of biodiesel from thraustochytrids was reconginzed
by international standards such as ASTM and EN14214 (Marchan Thraustochytrids are a promising future source for lipid
et al., 2017). compound production.
In addition to fatty acids, many thraustochytrids species with In order to obtain good strains, it is indispensable to disturb
high content of squalene or carotenoids have been isolated. or rewrite metabolic pathways. Fulfilling these targets needs a
Among the few squalene-producing thraustochytrid strains thorough understanding of lipid and terpenoid biosynthesis in
isolated to date, Aurantiochytrium can produce especially high thraustochytrids. Microbial lipid synthesis routes can be divided
titers, reaching more than 30% of DCW (Li et al., 2009). Although into fatty acid and TAG biosynthesis (Fig. 3). The PUFA
no impressive carotenoid yields was obtained in thraustochytrids, biosynthesis pathway of thraustochytrids is different to that of
experimental evidence shows that they have the potential to most oleaginous microorganism, which is the most critical reason
produce a greater variety of carotenoids including β-carotene, for it to efficiently produce DHA. Moreover, the terpenoid
astaxanthin and canthaxanthin (Aki et al., 2003). The biosynthetic mechanisms in thraustochytrids also has been
thraustochytrid strain AS4-A1 has the highest carotenoid content characterized. The following section provide an overview of the
reported to date, reaching up to 40 mg/g astaxanthin, which key biosynthetic pathways and metabolic engineering strategies.
corresponds to 4% of DCW (Benita et al., 2010). Notably, this 2.1. Biosynthesis of SFAs
thraustochytrid has a far higher carotenoid yield than the 6–7
mg/g obtained by the commercial production strain Phaffia In general, shorter fatty acids (< C18) with high content of SFA
rhodozyma and Haematococcus pluvialis (Schmidt et al., 2011). and monounsaturated fatty acid (MUFA) are preferable for
These production levels are only based on the optimization of biodiesel production because they increase the thermal and

Fig. 1. The fermentation process and function of lipid in thraustochytrids.


F. Du et al. Biotechnology Advances 48 (2021) 107725
oxidative stability of the resulting biodiesel (Gupta et al., 2015). in PKS enzymes successfully changed the chain lengths of the
Thraustochytrids can accumulate high amounts of SFA and MUFA resulting fatty acids (Fisch et al., 2011; Liu et al., 2014a). There
up to 76% of total fatty acids. It has proved that biodiesel derived are three PKS subunits in thraustochytrids, encoded by ORFA,
from thraustochytrids is comparable to fossil diesel in terms of ORFB, and ORFC. The ORFA protein contains one KS, one
energy usage and greenhouse gas emissions (Chang et al., acyltransferase (AT), a variable number of ACP, and one KR
2015b). SFAs are produced by the type I fatty acid synthetase domain. The number of ACP domains present in different
(FAS) pathway in thraustochytrids. In the first step, acetyl-CoA is thraustochytrids is different. For example, Aurantiochytrium sp.,
converted into malonyl-CoA by acetyl-CoA carboxylase (ACCase) Thraustochytrium sp., Schizochytrium sp. and Ulkenia sp. possess
(Fig. 3). A recent study found that the activity of ACCase was 13, 8, 9, and 10 ACPs, respectively (Fig. 5B). It has been reported
inhibited in Schizochytrium, but overexpression of an elongase that the number of ACPs is associated with the overall
enzyme for converting C16 into C18 fatty acids relieved the productivity (Lippmeier et al., 2009). When the number of ACPs
inhibition effect, resulting in elevated conversion of C15:0 into in Schizochytrium sp. was increased from 9 to 11, the DHA
C17:0 (Wang et al., 2019). Subsequently, malonyl-CoA is ligated productivity was enhanced by 80% (Hayashi et al., 2016). The
with ACP by MAT, after which a sequence encompassing ORFB protein contains one KS, one chain length factor (CLF), one
condensation by KS, reduction by KR, dehydration by DH, and AT, and one ER domain. The ORFC protein contains two DH and
repeated reduction by ER is performed by a multi- subunit one ER domain. Xie et al. (2017) advocated that the KS domain in
enzyme to yield C16 or C18 products (Parsons and Rock, 2013) ORFB is more related to PUFA biosynthesis, but the ORFA is more
(Fig. 4). Importantly, fatty acid chain extension is controlled by effective to improve SFA biosynthesis in Thraustochytrium sp.
acyltransferase and acyl-ACP thioesterase (TE). Notably, TE Recently, the control mechanism regulating carbon chain length
hydrolyzes acyl-ACP to release fatty acids, which is significant for in EPA and DHA biosynthesis was analyzed, and the results
the production of desired fatty acids. For example, when C12 and showed that the KS domain of ORFA controls the condensation
C14-specific TEs were overexpressed in Dunaliella tertiolecta, the from C18 to C20, while that of ORFC catalyze the condensation
content of C12:0 and C14:0 was increased 7- and 4-fold, from C20 to C22. Based on this discovery, microalgal DHA
respectively (Lin and Lee, 2017). There are two kinds of TEs, synthase was successfully converted to an EPA synthase (Dairi et
which have different preferences for fatty acids chain lengths al., 2019). Subsequently, the mechanism of double-bond
(Moreno-Perez et al., 2011´ ), suggesting that they can be used to formation in ARA and EPA biosynthesis was also clarified, in
improve lipid profile. 2.2. Biosynthesis of PUFAs which the DH domains of ORFB and ORFC play key roles in the
introduction of cis double bonds depending on the carbon chain
Two biosynthesis pathways of PUFAs are known to exist in length (Hayashi et al., 2019). When the DH domain of ORFC was
nature, the elongase-desaturase pathway and the polyketide-like disrupted, the PUFA yield of Schizochytrium sp. decreased by
synthase (PKS) pathway. Starting from SFAs, various desaturases more than half, highlighting its importance (Li et al., 2018a).
and elongases are required to produce a targeted PUFA, Taken together, research on the detailed biosynthetic pathways
proceeding via different intermediate PUFAs (Fig. 4). By contrast, and mechanisms controlling the specific PUFA profiles in
the PKS pathway catalyzes the de novo synthesis of PUFAs, in thraustochytrids is still in the preliminary stage, and further
which several cycles of reduction, dehydration, reduction and exploration is needed in the future.
condensation add two carbons in each cycle (Fig. 5A). There is a
controversy about the biosynthesis pathway of PUFAs in 2.3. Biosynthesis of TAGs
thraustochytrids. The Δ4 desaturase and Δ5 elongase were
detected in Thraustochytrium and Schizochytrium. Further fatty The oil of thraustochytrids contains neutral lipids (NLs),
acid analysis of two species showed that they had related phospholipids (PLs), glycolipids (GLs) and unsaponifiable matter
precursor fatty acids for DHA synthesis in the elongase- (UM) (Ren et al., 2014), with notable differences of the relative
desaturase pathway, which suggested that Thraustochytrium and proportions in different species. For example, NLs and PLs
Schizochytrium could synthesize DHA via the elongase-desaturase respectively account for 95% and 5% in Schizochytrium
pathway (Nagano et al., 2011). When the Δ12 desaturase gene of mangrovei (Fan et al., 2007), compared to 75% and 20% in
Thraustochytrium aureum was disrupted, the conversion of C18:1 Aurantiochytrium sp. (Liu et al., 2014b). The lipid fractions can be
into C18:2 decreased. However, the proportion of DHA increased, regulated by changing the cultivation conditions (Papanikolaou et
which suggested that DHA biosynthesis is independent of the al., 2010). Triacylglycerols (TAGs) are the major neutral storage
elongase-desaturase pathway (Sakaguchi et al., 2012). Hoang et lipids in thraustochytrids, while PLs are essential components of
al. (2016) supported that PKS and the elongase-desaturase the cell membrane. TAGs are synthesized via the Kennedy
pathway co-exist in Schizochytrium mangrovei PQ6 for PUFA pathway (Fig. 3). As the first step, glycerol-3-phosphate (G3P) is
biosynthesis. Altogether, different species of thraustochytrids converted into lysophosphatidate (LPA) by the rate-limiting
possessed different pathway of PUFAs biosynthesis: some via the enzyme glycerol-sn-3-phosphate acyl-transferase (GPAT).
elongase-desaturase pathway, some via PKS pathway, and some Strategies to increase lipid accumulation by enhancing GPAT
possess both pathways. expression have been successfully applied in a variety of
In fact, biosynthesis of PUFAs via PKS is more efficient microalgal species. Subsequently, lysophosphatidate acyl-
because it requires less NADPH (Figs. 4 and 5A). Using the transferase (LPAAT) transfers an acyl moiety from CoA onto the
elongase-desaturase pathway requires 26 NADPH to synthesize sn-2 position of LPA, resulting the phosphatidate (PA). Notably,
DHA from acetyl-CoA, while the PKS pathway only needs 14 LAPPT enzymes from different sources possess different substrate
NADPH molecules. Moreover, the PKS pathway produces fewer preferences, and an appropriate choice is a key for engineering
intermediate fatty acids due to fewer catalytic steps compared to strains with desired fatty acids (Balamurugan et al., 2017; Kim
the elongase-desaturase pathway. The PKS complex is composed and Huang, 2004). Finally, an acyl group is transferred to
of different subunits, and the number, location, and order of diacylglycerol from CoA under the catalysis of diacylglycerol
domains determine the finally produced fatty acids. For example, acyltransferase (DGAT). There are type-I and type-II DGAT,
the ARA-producing PKS possesses four subunits (Hayashi et al., whereby the latter more effective for TAG biosynthesis.
2019), while the EPA-producing PKS has three subunits and a Modification of DGAT genes has already been attempted and is
number of ACPs varying from four to three. Exchange of domains considered to be the best strategy for manipulating TAG
3
F. Du et al. Biotechnology Advances 48 (2021) 107725
accumulation in other microorganisms (Hsin-Ju et al., 2012; astaxanthin. Hong et al. (2013a) identified the squalene synthase
Irshad et al., 2015). However, this approach has not been applied of Aurantiochytrium sp. KRS101, which was the first evidence of
in thraustochytrids to date. the squalene biosynthesis pathway in thraustochytrids. Recently,
a top squalene producer named Aurantiochytrium sp. TWZ-97
2.4. Biosynthesis of terpenoids was isolated from coastal waters of China, with a squalene yield
of 188.6 mg/L. Seven key genes of the MVA pathway were
In addition to lipids, thraustochytrids can also be used as cell revealed by transcriptome analysis (Zhang et al., 2019). From the
factories for the production of terpenoids such as squalene and metabolic engineering point of view, squalene biosynthesis
carotenoids. shares the common precursor FPP with β-carotene, so
There are two common pathways for terpenoid biosynthesis in insufficient GGPP supply may be the reason for the lower
microorganisms - the mevalonate (MVA) pathway and the 2-C- carotenoid content. When a homologous enzyme from
methyl-D- erythritol-4-phosphate (MEP) pathway (Varela et al., Archaeoglobus with high capacity for GGPP synthesis was further
2015). It has been reported that terpenoid accumulation in overexpressed in Aurantiochytrium sp. SK4, the resulting
thraustochytrids proceeds via the MVA pathway (Fig. 3). engineered strain produced 78% more total carotenoids and
Terpenoid biosynthesis shares the common carbon precursors 5.02-fold more astaxanthin than the wild type (Ye et al., 2019). As
acetyl-CoA with lipid biosynthesis. Three molecules acetyl-CoA oleaginous microorganisms, thraustochytrids distribute the vast
are condensed into HMG-CoA by acetyl-CoA transferase (ERG10) majority of acetyl-CoA into the lipid biosynthesis pathway, thus
and HMG-CoA synthase (ERG13), which release two molecules of strategies for weakening lipid production as well as enhancing
CoA-SH. Next, HMG-CoA is reduced to mevalonate (MVA) by the MVA pathway could be used in the future. 3. Low-cost
HMG- CoA reductase (HMGR), and the reaction of which relies on culture medium
NADPH. Since this step is irreversible, HMGR is considered to be a
key rate- limiting enzyme in the MVA pathway. Phospho- Typical laboratory scale cultivation media for thraustochytrids
mevalonate kinase (ERG12) and mevanolate-5-P kinase (ERG8) is based on artificial seawater, containing glucose as carbon
are responsible for transferring a pyrophosphate group from ATP sources. However, in industrial fermentation of thraustochytrids,
to MVA-PP, accompanied by the release of ADP. Subsequently, the price of culture medium represents a significant proportion
mevanolate-5-PP decarboxylase (ERG19) and isopentenyl-PP of the operating costs. For example, in Schizochytrium sp.
isomerase (IDI) produce IPP and DMAPP, which are common fermentation for 120 h in 25L bioreactor, the cost of glucose
precursors for terpenoid synthesis. Next, farnesyl-PP synthase accounts for 73% of the total substrate cost (Fig. 2). Therefore,
(ERG20) condenses DMAPP and two molecules of IPP to generate low-cost culture medium such as crude glycerol, lignocellulosic
FPP, two molecules of which are utilized to form squalene by hydrolysate, and wastewater were tested in fermentation of
squalene synthase (Fig. 3). However, there are no reports on thraustochytrids (Table 1). However, these low-cost feedstocks
improving squalene production by direct genetic regulation of need further safety validation when seeking commercialization in
the MVA pathway in thraustochytrids. Starting from FPP, two and the field of food additives, for which it must be confirmed that
four steps are respectively needed to produce β-carotene and the product is safe for human consumption.

4
F. Du et al. Biotechnology Advances 48 (2021) 107725

Fig. 2. Substrate cost of Schizochytrium sp. fermentation in 25L bioreactor.


Table 1
Biomass accumulation and lipid content for thraustochytrids strain growth on low-cost carbon sources.
Strain Carbon source Biomass (g/L) Lipid content (% biomass) Fermentation (h) Reference

Aurantiochytrium sp. TC 20 glycerol 71 52% 69 Lee Chang et al., 2013


Thraustochytrium sp. BM2 glycerol 4.83 79% 48 Chen et al., 2020
Aurantiochytrium sp. glycerol 59.72 75.38% 168 Dilip et al., 2016
Schizochytrium sp. glycerol 54.15 72.38 168 Dilip et al., 2016
Aurantiochytrium limacinum SR21 Glucose and glycerol 88.32 83.84 96 Chang et al., 2015a
Aurantiochytrium sp. T66 glycerol 100 52 167 Jakobsen et al., 2008
Schizochytrium sp. S31 Glycerol 151.4 50.3 96 Chang et al., 2013b
Aurantiochytrium sp. FN21 sugarcane bagasse hydrolysate 33 35.9 120 Qi et al., 2017
Aurantiochytrium sp. T66 forest biomass hydrolysates 11.24 52.49 72 Patel et al., 2019
Schizochytrium limacinum SR21 sweet sorghum juice 9.4 73.4 96 Liang et al., 2010
Aurantiochytrium sp. KRS101 spent yeast 47.1 53.9 72 Ryu et al., 2013

5
F. Du et al. Biotechnology Advances 48 (2021) 107725

Fig. 3. Overview of lipid and terpenoid synthesis metabolism in thraustochytrids.


3.1. Glycerol acid cycle were significantly downregulated (Chen et al., 2016b).
Due to the generally poor regulation of glycerol assimilation, the
It has been reported that eukaryotic microorganisms conversion rate of glycerol into microbial oil is around 0.10 ± 0.02
dissimilate glycerol via phosphorylative pathway and g/g (Papanikolaou and Aggelis, 2011), lower than that of glucose.
dihydroxyacetone pathway (Wang et al., 2001). In Schizochytrium Similarly, the glycerol-to-oil conversion rate of thraustochytrids is
sp., glycerol is dissimilated via the phosphorylative pathway about 0.15 g/g (Chi et al., 2007; Pyle et al., 2008), indicating that
(Chang et al., 2013a). In this pathway, glycerol is converted into new strategies are needed to further improve the conversion rate
glycerol-3-phosphate (G-3-P) by glycerol kinase (GK), followed by in the future.
oxidation into dihydroxyacetone phosphate (DHAP) by FAD+-
dependent glycerol-3-phosphate dehydrogenase (FAD+-G-3- PDH)
(Makri et al., 2010). Previous studies had shown that lipid
accumulation by thraustochytrids grown on glycerol was
comparable with those grown on glucose (Chi et al., 2007).
Jakobsen et al., (2008) produced 90–100 g/L DCW containing 54–
63% of lipids by culturing Aurantiochytrium sp. on glycerol.
Combined with the highest kLa, the DCW and lipid concentration
of Schizochytrium sp. S31 can reach up to 151.4 g/L and 79.7 g/L,
respectively (Chang et al., 2013a). When 10 g/L glycerol was used
as carbon source and 12.5 g/L corn steep liquor as nitrogen
source, the maximal biomass of Thraustochytrium sp. BM2
reached 4.83 g/L, containing 79% of lipids, suggesting that this
strain could serve as a low-cost heterotrophic lipid producer
(Chen et al., 2020). However, the activity of the key enzymes GK
and G-3-PDH involved in the glycerol assimilation pathway may
be inhibited due to decreased oxygen supply at the later
fermentation stage. Therefore, mixed carbon sources with
glucose and glycerol were used to culture Thraustochytrids,
resulting maximal growth and DHA productivity (Li et al., 2015, Ye
et al., 2020). Moreover, the addition of calcium and magnesium
can also address these problems, because calcium can enhance
the activity of G-3-PDH and magnesium promote the activity of
malic enzyme in thraustochytrids (Dilip et al., 2016). Interestingly,
although the amounts of biomass and lipids of thraustochytrids
were similar in the cultivation with glucose or glycerol, the DHA
content was enhanced when glycerol was used as the carbon
source. Subsequent transcriptomic analysis revealed that acetyl-
CoA production was overall increased because the genes involved
in glycolysis and amino acid metabolism were remarkably
upregulated, while the pentose phosphate pathway and citric

6
F. Du et al. Biotechnology Advances 48 (2021) 107725
with the ability to grow on carboxymethylcellulose, a β-
glucosidase from Aspergillus aculeatus was expressed in
Aurantiochytrium sp., and the resulting strain with cellobiose as
the sole carbon source exhibited increased growth and enzyme
activity.
The process of lignocellulose pretreatment inevitably yields
many toxic by-products, which can inhibit cell growth. Therefore,
an Aurantiochytrium sp. strain tolerant to lignocellulosic
hydrolysate was isolated and commercially applied for DHA
production (Huang et al., 2008). Furthermore, high-throughput
RNA sequencing was applied to clarify the tolerance mechanism,
and results showed that genes involved in the TCA cycle and
amino acid biosynthesis were highly upregulated, while genes
involved in the degradation of aromatic compounds were
downregulated (Qi et al., 2017). It should be noted that different
species of thraustochytrids vary greatly in their xylose utilization
capacity, and the mechanisms of hydrolysate tolerance should be
studied further.

3.3. Wastewater

Microbial fermentation often results in large amounts of


fermentation wastewater. For instance, the fermentation of
Aurantiochytrium generates 100,000 tons of wastewater every
year in China (Chen et al., 2016a). Although wastewater
treatment is not expensive, it requires high-power processing
equipment (Ruiz-Rosa et al., 2016). The fermentation wastewater
contains inorganic salts or growth-promoting factors, which can
potentially be valuable for the fermentation of thraustochytrids.
For example, the supernatant of distillery waste contains 2.5% of
proteins as well as 0.2% of amino acids, and has been used as the
sole nitrogen source for lipid and astaxanthin production using
Schizochytrium sp. (Yamasaki et al., 2006). The fermentation
Fig. 4. The fatty acid synthetase (FAS) pathway and the elongase- wastewater from Mortierella alpina was also used to replace
pure water for Aurantiochytrium fermentation, resulting in a DHA
desaturase pathway. 3.2. Lignocellulosic hydrolysate
yield of 30.4 g/L, with no significant changes compared to the
use of pure water (Song et al., 2017). In addition, it has been
The use of lignocellulosic biomass is expected to increase
demonstrated that saline wastewater from cheesemaking
significantly due to which is the most abundant renewable
(Humhal et al., 2017), industrial wastewaters from pig
organic materials. After pretreatment, sugars including glucose,
slaughtering plants (Villarroel Hipp and Silva Rodriguez, 2018),
xylose, and arabinose can be obtained and then be utilized as
K2HPO4-waste feeds (Leong et al., 2019), and recycled spent
carbon sources by microorganisms. There are two xylose-
media (Bagul and Annapure, 2020) can be used to lower the
utilization pathways in microorganisms, the eukaryotic xylose
production cost of thraustochytrid biomass and lipids. In general,
reductase/xylitol dehydrogenase pathway and prokaryotic xylose
wastewater cannot be used directly because it contains cell
isomerase pathway (Schellenberg et al., 1984; Xin et al., 2014).
growth inhibitors, and it would be highly desirable to optimize
Compared to the xylose isomerase pathway, the xylose
the handling methods and enhance the productivity of
reductase/xylitol dehydrogenase pathway has more catalytic
wastewater fermentations in the future. Overall, using
steps, and the formation of xylitol affects the conversion rate of
wastewater for the production of biomass and value-added
xylose. In addition, heterologous expression of the xylose
products is in high demand.
reductase/xylitol dehydrogenase pathway can disturb the
intracellular cofactor balance. It should be noted that different
4. Molecular approaches for enhancing lipid and terpenoid
species of thraustochytrids vary greatly in their xylose utilization production
capacity. Schizochytrium sp. can consume xylose from sugarcane
bagasse hydrolysate (Hoang et al., 2018). By using synthetic The genome sequences of thraustochytrids such as
biology techniques, the ability of microorganisms to metabolize Schizochytrium sp., Aurantiochytrium limacinum SR21 and
xylose can be enhanced. Thraustochytrids produce xylitol from Schizochytrium aggregatum have been published. In recent
both glucose and xylose, but cannot grow in the presence of years, a transformation method for genome editing has been
xylose alone due to an incomplete xylose reductase/xylitol successfully established in thraustochytrids. Although the
dehydrogenase pathway. When introduced endogenous xylose efficiency is still low, metabolic engineering of thraustochytrids to
isomerase and heterologous xylulose kinase into thraustochytrid, improve the accumulation of target products is gradually
the resulting strain can utilize xylose ( (Merkx et al., 2018)). When becoming more feasible (Table 2).
Aurantiochytrium sp. was cultivated using birch hydrolysate
pretreated with organic solvents, it resulted in 703.3 mg/L/ day
of DHA productivity and 1.0 g/L of squalene, which suggests
economic production of lipid compounds is technically feasible
(Patel et al., 2019). Furthermore, in order to endow the strain

7
F. Du et al. Biotechnology Advances 48 (2021) 107725
4.1. Genetic engineering

8
F. Du et al. Biotechnology Advances 48 (2021) 107725
Selection markers are genes that are introduced into a cell to
allow its survival under predetermined selection conditions. In
thraustochytrids, selectable markers such as resistance genes for
zeocin, G418, hygromycin, cycloheximide, and paromomycin can
be used (Sakaguchi et al., 2012; Suen et al., 2014; Hong et al.,
2013a; Bayne et al., 2013). In most studies, electroporation with
linear DNA is the most common strategy for transformation (Li et
al., 2018b; Wang et al., 2019). In addition, particle bombardment
and Agrobacterium-mediated transformation can also be used in
thraustochytrids (Cheng et al., 2012). In order to make the

Fig. 5. A: Catalytic steps of the PKS pathway in thraustochytrids. B: Structure of PKS pathway in different thraustochytrids species.
transformants suitable for industrial application, the cre/loxP
system was established to generate markerless mutations in A.
limacinum (Sun et al., 2015). Homologous recombination has
been shown to be effective for integrating a foreign gene into the
genome, but the required length of the homology arms is
generally 1000-2000 bp. Recently, a 2A peptide-based method
was developed in Aurantiochytrium for multiple genes expression
(Wang et al., 2020).

9
F. Du et al. Biotechnology Advances 48 (2021) 107725
At present, there is little research improved lipid production modifying the PKS pathway, which may mainly due to the lack of
by modifying the FAS pathway. Recently, the record lipid yield of efficient genetic tools, together with the complexity and unclear
110.5 g/L was obtained by overexpressing MAT in function of PKS enzymes.
Schizochytrium, leading to 39.6% higher production than in the In addition, many studies aimed to enhance the performance
wild-type strain (Li et al., 2018b). When ACCase was of thraustochytrids by regulating the bypass pathway. It is
overexpressed in Schizochytrium using the strong constitutive generally accepted that lipid biosynthesis requires large amounts
ccg1p promoter, the lipid content and DHA yield respectively of acetyl-CoA and NADPH as the main precursor and source of
increased by 30.5 and 41.9% (Han et al., 2020). Several metabolic reducing power, respectively. In Aurantiochytrium sp., the acetyl-
engineering studies focused on the PUFA biosynthesis pathway. CoA synthetase gene from Escherichia coli was introduced to
Such approaches can be used to increase the content of naturally increase the pool of acetyl-CoA, which resulted in a maximal fatty
occurring fatty acids that are present at low levels in the wild acid content of 46.8% (Yan et al., 2012). More significantly,
type strain. For example, when a Δ5 desaturase from overexpression of acetyl-CoA synthetase in Schizochytrium not
Thraustochytrium aureum was overexpressed in A. limacinum, only improved carbon source utilization, but also decreased the
the levels of EPA and ARA were respectively increased 4.6- and production of the major byproduct acetate (Yan et al., 2012).
13.6- fold in the transgenic strain (Hauvermale et al., 2006). Based on β-galactosidase reporter system, a strong promoter of
Disruption of the Δ12 desaturase of Thraustochytrium aureum ccg1p was screened, then ATP-citrate lyase and ACCase under
ATCC 34304 resulted in the accumulation of oleic acid (C18:1) ccg1p were overexpressed in Schizochytrium sp. ATCC 20888,
and a decrease of linoleic acid (C18:2) (Takanori et al., 2012). In resulting in the highest lipid content of 73% (Han et al., 2020).
our previous study, the heterogeneous ω-3 desaturase of The NADPH needed for fatty acid synthesis is mainly produced by
Saprolegnia diclina was expressed in Schizochytrium sp., the the transhydrogenase cycle and the PPP pathway, in which the
result showed that more DPA was transformed into DHA (Re et ME and the G6PDH is the key enzymes involved in two pathways,
al., 2015). While ω-3 desaturases possess an extensive substrate respectively. Cui et al. (2016) overexpressed the G6PDH for
specificity, the ω-3 desaturase from Saprolegnia diclina was increasing NADPH supply via the pentose phosphate pathway,
shown to be more specific for C20:4 (Pereira et al., 2004). Thus, and the PUFAs content was increased by 10.6%. Similarly, the
protein engineering of the ω-3 desaturase may be a feasible NADPH generation system of Aurantiochytrium sp. was
strategy to improve its DPA specificity. strengthen by overexpressing the gene encoding malic enzyme,
In theory, rewriting the PKS pathway is the most direct and the recombinant strain exhibited a high SFA yield of 8.65 g/L
method to regulate PUFA biosynthesis. ORFA from (Cui et al., 2019).
Thraustochytrium contains 8
ACPs, while that of Schizochytrium possesses 9. The main role
4.2. Chemical activators
of ACP domains is to covalently bind the acyl substrates. It has
been reported that the amount of ACPs influences PUFA
Many small-molecule drugs can be used to stimulate or
productivity. In order to
Table 2
Genetic engineering strategies to improve lipid accumulation in thraustochytrids.
Strain Transformation Strategy Performance Reference

Schizochytrium sp. TIO1101 Electroporation transformation (2100 Overexpression of ACS gene from The biomass and fatty acid proportion of transformants Yan et al., 2012
V, 4.5 ms) Escherichia coli was increased by 29.9 and 11.3%, respectively.

Thraustochytrium aureum Electroporation transformation Disruption of △5 desaturase gene The content of dihomo-γ-linolenic acid (C20:3) and Sakaguchi et
ATCC 34304 (50 μF, 50 Ω, 7.5 kV/cm, 2 times) eicosatetraenoic acid (C20:4) was increased. al., 2012
Schizochytrium Electroporation transformation Overexpression of malonyl-CoA: ACP The DHA and EPA yield was enhanced by 81.5 and Li et al., 2018b
transacylase (MAT) 172.5% than that of wild type, respectively.
Thraustochytrium aureum Electroporation transformation Disruption of △12 desaturase gene The oleic acid content was increased and linoleic acid Takanori et al.,
ATCC 34304 was decreased. 2012
Schizochytrium sp Electroporation transformation Overexpression of G6PDH The PUFA was increased by 10.6% Cui et al., 2016
Schizochytrium sp Electroporation transformation (0.75 Introduction of ω-3 desaturase gene from The 3% (DPA) was converted to (DHA) Re et al., 2015
kV, 200 Ω, and 50 μF) Saprolegnia diclina
Aurantiochytrium sp. SK4 Electroporation transformation (1.8 Expression of vitreoscilla hemoglobin The fatty acid and astaxanthin content of Suen et al., 2014
kV, 300 Ω, and 50 μF) transformants was 44% and 9-fold higher than that of
the wild type
Schizochytrium sp Electroporation transformation (0.75 Replacement of the native AT with AT The EPA content was increased by 3.7 times. Ren et al., 2018
kV, 200 Ω, and 50 μF) domain from Shewanella sp.
Schizochytrium sp. ATCC Electroporation transformation (0.75 Overexpression of ATP-citrate lyase The DHA yield was increased by 48.8%. Han et al., 2020
20888 kV, 200 Ω, and 50 μF) (ACL) and acetyl-CoA carboxylase
(ACCase)
Aurantiochytrium Electroporation transformation Expression of the EPA biosynthetic gene The EPA yield was increased by 5-folds. Wang et al.,
cluster from Shewanella japonica 2020

examine their function, the number of ACPs in Schizochytrium inhibit intracellular metabolic pathways, which are equivalent to
was increased from 9 to 10 and 11. The results showed that the effects of overexpression and suppression approaches. Compared
PUFA yield of the strain with more ACP domains is 50 to 100% to genetic engineering, the application of chemicals is potentially
higher than that of the native group (Hayashi et al., 2016). The AT an easy and practical approach to improve lipid production in
domain of Schizochytrium sp. was replaced with the AT domain thraustochytrids, which has become one of research hotspots.
from Shewanella sp., which resulted in a 3.7 times increase of Phytohormones and their derivatives are often used to improve
EPA production compared to the wild type (Ren et al., 2018). lipid accumulation by adjusting the internal biochemical
Furthermore, in a recent study, the whole EPA biosynthesis gene pathways. For example, lipid accumulation of Schizochytrium was
cluster from Shewanella japonica was expressed in enhanced to 16.79% by adding 20 mg/L jasmonic acid and 2
Aurantiochytrium, resulting in 5-fold increase of EPA yield (Wang mg/L naphthoxyacetic acid (Wang et al., 2018a). Similarly, the
et al., 2020). To date, no major progress has been made in addition of 6-benzylaminopurine increased the lipid yield of
10
F. Du et al. Biotechnology Advances 48 (2021) 107725
Aurantiochytrium sp. by 25.9% (Yu et al., 2016). In a recent study, supply is a classic and effective control strategy for
the addition of 120 mg/L inositol improved the production of thraustochytrid fermentations, since DHA is produced by the
lipids and DHA in Schizochytrium sp. by 13.9 and 20.8%, oxygen-independent PKS pathway. For example, stepwise
respectively (Liu et al., 2019). Moreover, short-chain alcohols can dissolved oxygen control was applied to thraustochytrids, with a
partition into membranes to increase their fluidity and interfere high DO stage for cell growth and low DO stage for lipid
with normal cell functions, which was also used to regulate the accumulation. Moreover, when the DO level was maintained at
accumulation of terpenoids. A remarkable 2000-fold increase of 50% in the whole fermentation process, Aurantiochytrium
astaxanthin was obtained in Schizochytrium limacinum by adding limacinum SR21 produced DCW at 8.82 g/L/day and DHA
5.6% methanol (Du et al., 2019). Interestingly, addition of 6 g/L productivity at 2.9 g/L/day (Huang et al., 2012). However, it has
butanol in the same species decreased the DHA content by been shown that DO does not fully reflect the oxygen supply
nearly 40%, but the squalene content was increased 31 times capacity, especially under DO-limited conditions (Chang et al.,
(Zhang et al., 2017). 2013b). Consequently, the oxygen uptake rate (OUR) was
Sterol biosynthesis shares acetyl-CoA as a common precursor introduced as an online parameter to better reflect the oxygen
with the fatty acid and terpenoid biosynthesis pathways, so that supply in fermentations of Schizochytrium sp. (Chang et al.,
inhibiting the sterol biosynthesis pathway could be an effective 2014). In addition, Guo et al. (2016) combined the OUR and
strategy to improve the production of the former two types of respiratory quotient (RQ) as the real-time monitoring
products. Li et al. (2019a) used fluconazole to block the sterol parameters, results showed that this method achieved the
pathway by inhibiting lanosterol 14α- demethylase activity, which highest DHA content (45% in total fatty acids), suggesting that it
resulted in a 16 and 45% increases of the lipid and squalene is more effective than DO in Schizochytrium sp. fermentation.
content in Schizochytrium, respectively. In addition, Shirasaka et Nutrient limitation is another effective strategy for lipid
al. (2005) found that benzoic acid enhanced EPA accumulation in production in thraustochytrids. For example, lipid production of
Schizochytrium sp. by suppressing the methyl-directed Aurantiochytrium sp. T66 was significantly boosted when the
desaturation from the Δ13 position. Based on this, four benzoic cells were subjected to nitrogen limitation (Jakobsen et al., 2008).
acid derivatives were tested in Schizochytrium limacinum SR21, Moreover, nitrogen limitation is conducive to neutral lipids
and the addition of 200 mg/L of p-aminobenzoic acid increased accumulation but nor polar lipids in Schizochytrium sp. (Sun et
the lipid yield by 56.84% (Li et al., 2019b). In order to alleviate al., 2014). However, temperature is more effective for regulating
lipid peroxidation, which is especially important for PUFAs, fatty acid composition. At lower temperatures, more PUFAs need
antioxidants can been used to scavenge intracellular reactive to be synthesized to maintain the fluidity of the cell membrane
oxygen species. When 9 g/L of ascorbic acid was added to (An et al., 2013). Accordingly, when the temperature was
cultures of Schizochytrium sp., the DCW and DHA yield decreased to 12 ◦C from 30 ◦C, the DHA content of
respectively increased by 16.16 and 30.44%, which was Aurantiochytrium mangrovei Sk-02 was increased from 29 to 42%
accompanied by lower ROS levels and higher antioxidant capacity in the late lipid accumulation phase (Chodchoey and Verduyn,
(Ren et al., 2017). In addition, supplementing butylated 2012). However, this strategy could less feasible in industrial
hydroxytoluene or propyl gallate also improved the lipid yield and production due to the increased cost of lower temperature
cell growth of thraustochytrids (Singh et al., 2015). Overall, the control. To overcome this bottlenecks, Sun et al. (2018) applied
addition of chemicals is a sustainable and economical strategy adaptive evolution under low temperature to enhance the PUFA
that can be easily applied in large-scale production. content of Schizochytrium sp., which avoid the high cost and
5. Engineering operation strategies for enhancing lipid and production instability. In a recent study, ammonia and citric acid
terpenoid production was applied as pH regulators, then a two-stage pH control
strategy was developed for Schizochytrium sp. fermentation,
The lipid production in thraustochytrids is often influenced by resulting a maximum DHA yield of 272.92 mg/L/h (Yin et al.,
the culture environment, which exhibit two opposing 2018b). Altogether, many bioprocess engineering strategies have
characteristics, including high biomass with low lipid content, or been developed to improve lipid accumulation of
low biomass with high lipid content. Thus, many operation thraustochytrids, with varying degrees of success.
strategies were developed to balance the lipid accumulation and
cell growth (Table 3). In addition, it is difficult to enlarge the 5.2. Bioprocess scale-up strategies
production quantities from the laboratory-scale while keeping
the titers similar, thus scale-up technologies and bioreactor The aim of scale-up is to enlarge the production quantities
design are key to industrialization. while keeping the titers similar to those obtained on the
laboratory-scale. The oxygen supply conditions obviously affect
cell growth and lipid production, so the volumetric oxygen-
5.1. Two-stage cultivation strategies transfer coefficient (KLa) is often chosen as the critical factor for
scale-up. Using this scale-up strategy, the fermentation
Thraustochytrids cells accumulate lipid as an energy storage performance of Schizochytrium sp. in a 7000L stirred-tank
battery when exposed to a stress condition, but which could bioreactor (DHA content of 19.72 g/L) was similar to that of the
generate growth limitation. Two-stage strategy is an effective experimental-scale process (DHA content of 13.84 g/L), but it
cultivation system due to which separate the lipid accumulation resulted in
phase from the biomass growth phase. Regulating the oxygen
Table 3
Operation strategies for high-cell-density fermentation.
Strain Strategies Biomass Lipid DHA References
(g/L) yield yield
(g/L) (g/L)

11
F. Du et al. Biotechnology Advances 48 (2021) 107725
Schizochytrium sp. The Volumetric 151.40 76.15 28.93 Chang
S31 mass transfer et al.,
coefficient (kLa) 2013b
was maintained
at
1802 ± 105 h
−1
Schizochytrium sp. Two-phase pH 120.6 62.6 32.8 Yin et al.,
control with 2018b
ammonia and
citric acid
Schizochytrium sp. The fluid dynamics 128.3 74.8 39.2 Guo et al.,
was analyzed for 2018
improve the
efficiency of scale-
up, and and
impeller
combinations was
optimized.

Schizochytrium sp. Combined porous- 151.0 103.6 44.3 Guo et al.,


membrane- 2017
impeller
bioreactor and
three-stage DO
control strategy

Aurantiochytrium sp. Limitation of 100 58 11.84 Jakobsen et


T66 nitrogen and al.,
oxyen 2008
a 150-fold quantitative scale-up (Qu et al., 2013). Furthermore, Guo et al. (2018) analyzed the fluid dynamics of different scale
bioreactors and then developed the efficient and economical scale-up strategy for DHA production using Schizochytrium sp., which
resulted in 39.2 g/L DHA yield in 7000L bioreactor. In addition, feeding strategy is also an important factor in scale-up production. Using
continuous feeding strategy with a glucose concentration of 20–25 g/L achieved a maximum DHA productivity of 320.17 mg/L/ ⋅h in
500-L pilot-scale bioreactor, which represent increase of 37.68% over that of the intermittent feeding method (Guo et al., 2020).

5.3. Bioreactor design

In the fermentation process of thraustochytrids, new reactor designs need to provide an adequate oxygen supply but avoid high
shear forces, so air-lift bioreactors is a better choice. When an air-lift fermenter was used to culture Aurantiochytrium sp., the DHA
content was increased from 39.5 to 52.3% (Hong et al., 2013b). The highest DCW of 27.57 g/L and DHA productivity of 610 mg/L/day
were achieved in an air-lift fermenter, due to the low shear stress (Chen and Yang, 2018). Furthermore, the impeller configuration was
optimized to improve the lipid production of Schizochytrium sp. The combination of two upper 6- SBDTs and one bottom 6-ABDT was
proven to be highly beneficial for DHA synthesis (Zhao et al., 2016). Guo et al. (2018) applied an impeller configuration with two wide-
blade hydrofoil impellers pumping downward (located at the top and in the middle), and a six-parabolic-blade disk turbine (located at
the bottom) in a 7000L bioreactor. This configuration resulted in a DHA concentration of 326.67 mg/L/h, representing an increase of
133.3% over the original impeller. In addition, membrane materials are also widely used in new reactor design. For example, a hollow-
fiber polypropylene membrane was mounted in a
shake flask to transform it into a membrane reactor, which led to to the specificity and complexity of cell walls, this method may
a 60% relative increase of DHA productivity in Schizochytrium sp. require screening of wall- breaking enzymes for each species.
compared to single shake mode (Zhang et al., 2013). An Here, we mainly discuss lipid extraction technologies and the
innovative porous- membrane-impeller bioreactor was recently reuse of algal residues.
designed, and it was found to significantly facilitate cell
proliferation and DHA accumulation in Schizochytrium sp. when 6.1. Lipid extraction
combined with a multi-stage control strategy (Guo et al., 2017).
Lipid extraction is a crucial stage in industrial lipid production,
6. New developments in downstream processing aiming to facilitate the release of lipids from cells. Organic
solvent extraction is the most commonly used method in
The key stages of downstream processing include cell thraustochytrids, and the suitability of the organic solvent
harvesting and disruption, lipid extraction, and conversion of determines the extraction efficiency. Byreddy et al. (2015) tested
biomass into products (Fig. 1). Centrifugation has been suggested nine organic solvents for lipid extraction from thraustochytrids,
as the most effective cell harvesting method for thraustochytrids, and the results showed that hexane generated the highest lipid
due to advantages such as speed, efficiency, low cost, no toxic content of 12.5%. Furthermore, two or more organic solvents can
residues, and not affecting the quality of the biomass. Common be used simultaneously in polar/non-polar combinations to
cell disruption techniques for thraustochytrids include osmotic increase the lipid yield. The maximum lipid extraction yield from
shock, grinding, high-pressure homogenizers and other thraustochytrids was 22% using a chloroform: methanol ratio of
mechanical approaches. In contrast, enzymatic treatment for 2:1 (Byreddy et al., 2015). Organic solvent extraction is simple in
lipid release is a low-energy and environmentally friendly operation and has a high efficiency, but it leaves an unavoidable
process, which mainly includes the disruption of cell wall and solvent residue, which is unacceptable for many product
internal organelles for free lipid bodies releasing. However, due applications. In addition, organic solvent extraction can cause

12
F. Du et al. Biotechnology Advances 48 (2021) 107725
environmental pollution and affect the health of operators 7. Conclusions
(Harris et al., 2018). Ionic liquids have been shown to improve
the efficiency of organic solvent extraction of lipids under mild Thraustochytrids are a promising and competitive source of
conditions. The two ionic liquids imidazolium 1-ethyl- 3- lipid production compared to other microorganisms. Its high
methylimidazolium ethylsulfate [C2mim] [EtSO4] and growth rate, broad substrate utilization, efficient product
tetrabutylphosphonium propanoate [P4444] [Prop] were pathways, and mature operation technologies, in conjunction
evaluated in lipid extraction from Thraustochytrium sp., and the with compact downstream processing, all point to great promise
results indicated that both ionic liquids facilitated the lipid for the future development of thraustochytrids. On one hand, the
extraction (Zhang et al., 2018). Furthermore, supercritical fluids fatty acid composition of thraustochytrids is simple, dominated
can be used to replace organic solvents in extraction processes. by C16:0 and DHA. Whereas DHA are used in the field of human
For instance, a lipid yield of 33.9% with a DHA content of 27.5% health, C16:0 is suitable for biodiesel production. Based on
was obtained from Schizochytrium limacinum using supercritical precursor of acetyl-CoA, C16:0 and DHA are produced by two
CO2 (Tang et al., 2011). In a recent study, a novel method for lipid different enzymatic systems, therefore it is possible to favor one
extraction from thraustochytrids was developed. The biomass pathway over another by genetic engineering. In addition, the
was first combined with potassium hydroxide, after which PKS also generates about 10% of C22:5, so it is meaningful to
reaction mixture was added to an anti-solvent to generate solid convert DPA into DHA via introducing w-3 desaturase. On the
precipitates, and the final PUFA recovery rate reached 72–74% other hand, the efficient PKS pathway is the key value of
(Pour et al., 2020). It is worth noting that thraustochytrid lipids thraustochytrids, so how to excavate and use this pathway needs
contain a series of fatty acids, mainly including C16:0, C22:5 and to be considered in the future. In fact, the FAS pathways has been
C22:6. Since the lipid bodies are large and insoluble in water, it is engineered to achieve desired fatty acids by in vitro and in vivo
possible to extract lipids by simple centrifugation. Three-phase assays, which encourage that similar approaches could be taken
centrifuges can separate the lipids, water and solids, and this to modify the PKS pathway for producing other high-value fatty
simple technology is used in industrial lipid extraction from acids such as EPA. There is also considerable potential for the
Schizochytrium sp. production of more high-valued products using thraustochytrids.
In other words, it would be interesting to convert
6.2. Residue application thraustochytrids from DHA producer to other chemical producer
in the future, in which, squalene and carotenoids are surely good
Microalgal biomass residues are major by-products of biodiesel examples. However, resources and tools used for genome editing
production, and can potentially be utilized as nutrient sources remains limited the development of thraustochytrids. A primary
for microbial cultivation due to their content of proteins and challenge is the lack of the key enzyme information involved in
carbohydrates. For instance, fermentation residues were terpenoids biosynthesis. Thus, developing efficient genetic
hydrolyzed into amino acids and sugars for Chlorella vulgaris editing tools and discovering molecular targets is therefore of
cultivation, which yielded 3.28 g/L of biomass with a lipid much significance.
content of 35% (Zheng et al., 2012). It has been reported that
Schizochytrium sp. contains 3.45% of total nitrogen (Yin et al., Declaration of Competing Interest
2018a), and its oil-extraction residues can potentially be
developed into a novel nitrogen source (Bryant et al., 2012). In The authors declare that they have no competing interests.
order to increase the economic and environmental sustainability Acknowledgement
of Schizochytrium sp. fermentation, its residues were used to
replace yeast extract for its own fermentation (Yin et al., 2018a). This work was supported by the Nature Science Foundation of
A 27.1 g/L of DHA yield was obtained when 80% of yeast extract
Jiangsu Province (No. BK20190706, No. BK20170988), the
nitrogen was replaced with the residues, which was 20.07%
higher than that of the control. National Natural Science Foundation of China (Number:
In addition, residues of thraustochytrid fermentation are rich 21908112, No. 22038007). References
in PUFAs and proteins that are beneficial to animal growth, so
Aki, T., Hachida, K., Yoshinaga, M., Katai, Y., Yamasaki, T., Kawamoto, S.,
which are widely used as an ingredient of livestock feed (Bruneel Kakizono, T., Maoka, T., Shigeta, S., Suzuki, O., Ono, K., 2003.
et al., 2013). For example, the EPA and DHA content of eggs was Thraustochytrid as a potential source of carotenoids. J. Am. Oil Chem. Soc.
significantly increased by feeding chickens with Aurantiochytrium https://doi.org/10.1007/s11746-003- 0773-2.
An, M.L., Mou, S.L., Zhang, X.W., Ye, N.H., Zheng, Z., Cao, S.N., Xu, D., Fan, X.,
biomass residues, thus improving the nutritional and economic Wang, Y. T., Miao, J.L., 2013. Temperature regulates fatty acid desaturases
value of eggs (Moran et al., 2020). Moreover, numerous studies at a transcriptional level and modulates the fatty acid profile in the
indicate that enhancing the DHA content in feed can improve the Antarctic microalga Chlamydomonas sp. ICE-L. Bioresour. Technol.
https://doi.org/10.1016/j.biortech.2013.01.142.
survival rate of aquatic animals (Ganuza et al., 2008; Gładkowski
Ana, M.D.O.F., Luis, D.G.M., Júlio, C.D.C., Gilberto, V,D,M,P., Vanete, T,S., Carlos,
et al., 2014). Fermentation residues of thraustochytrids have R,S., 2016. Technological trends and market perspectives for production of
been used as feed additives for a variety of aquatic animals, microbial oils rich in omega-3. Crit. Rev. Biotechnol.
including atlantic salmon parr and pacific white shrimp (Miller et https://doi.org/10.1080/ 07388551.2016.1213221.
Bagul, V.P., Annapure, U.S., 2020. Effect of sequential recycling of spent media
al., 2007). When Schizochytrium biomass residue was added to wastewater on docosahexaenoic acid production by newly isolated strain
pig feed, the serum triglycerides of the pigs significantly Aurantiochytrium sp. ICTFD5. Bioresour. Technol. https://doi.org/10.1016/j.
decreased, and the DHA content in subcutaneous fat increased biortech.2020.123153.
Balamurugan, S., Wang, X., Wang, H.L., An, C.J., Li, H., Li, D.W., Yang, W.D., Liu,
13 times (Jon Meadus et al., 2011). These studies indicate that
J.S., Li, H.Y., 2017. Occurrence of plastidial triacylglycerol synthesis and the
feeding the fermentation residues of thraustochytrids can have potential regulatory role of AGPAT in the model diatom Phaeodactylum
unique positive effects on the physiological function of livestock, tricornutum.
improving animal growth and product quality. Biotechnol. Biofuels. https://doi.org/10.1186/s13068-017-0786-0.
Bayne, A.C.V., Boltz, D., Owen, C., Betz, Y., Maia, G., Azadi, P., Archer, H.S., Zirkle,
R., Lippmeier, J.C., 2013. Vaccination against influenza with recombinant
hemagglutinin expressed by Schizochytrium sp. confers protective
immunity. PLoS One. https://doi.org/10.1371/journal.pone.0061790.

13
F. Du et al. Biotechnology Advances 48 (2021) 107725
Benita, Q., Ivonne, H., Emilio, H. Andr´es, 2010. Docosahexaenoic acid (C22: 6n− carbon- chain length in polyunsaturated fatty-acid synthases. Angew.
3, DHA) and astaxanthin production by Thraustochytriidae sp. AS4-A1 a Chem. Int. Edit.
native strain with high similitude to Ulkenia sp.: evaluation of liquid https://doi.org/10.1002/anie.201900771.
residues from food industry as nutrient sources. Enzyme. Microb. Tech. Dilip, Singh, Barrow, Colin J., Munish, Puri, Tuli, Deepak K., Mathur, Anshu S.,
https://doi.org/10.1016/j. 2016. Combination of calcium and magnesium ions prevents substrate
enzmictec.2010.04.002. inhibition and promotes biomass and lipid production in thraustochytrids
Bruneel, C., Lemahieu, C., Fraeye, I., Ryckebosch, E., Muylaert, K., Buyse, J., under higher glycerol concentration. Algal Res.
Foubert, I., 2013. Impact of microalgal feed supplementation on omega-3 https://doi.org/10.1016/j.algal.2016.02.024.
fatty acid enrichment of hen eggs. J. Funct. Foods. Doi, K., Honda, D., 2017. Proposal of Monorhizochytrium globosum gen. nov.,
https://doi.org/10.1016/j.jff.2013.01.039. comb. nov. (Stramenopiles, Labyrinthulomycetes) for former
Bryant, H.L., Gogichaishvili, I., Anderson, D., Richardson, J.W., Sawyer, J., Thraustochytrium globosum based on morphological features and
Wickersham, T., Drewery, M.L., 2012. The value of post-extracted algae phylogenetic relationships. Phycol. Res. https://doi.org/
residue. 10.1111/pre.12175.
Algal Res. https://doi.org/10.1016/j.algal.2012.06.001. Du, H., Liao, X., Gao, Z., Li, Y., Lei, Y., Chen, W., Chen, L., Fan, X., Zhang, K., Chen,
Byreddy, A.R., Gupta, A., Barrow, C.J., Puri, M., 2015. Comparison of cell S., 2019. Effects of methanol on biomass, and fatty acid and carotenoid
disruption methods for improving lipid extraction from Thraustochytrid biosynthesis in Schizochytrium limacinum B4D1. Appl. Environ. Microbiol.
strains. Marine Drugs. https://doi.org/10.3390/md13085111. https://doi.org/10.1128/ AEM.01243-19.
Chang, G., Luo, Z., Gu, S., Wu, Q., Chang, M., Wang, X., 2013a. Fatty acid shifts Fan, K.W., Jiang, Y., Faan, Y.W., Chen, F., 2007. Lipid characterization of mangrove
and metabolic activity changes of Schizochytrium sp. S31 cultured on thraustochytrid-Schizochytrium mangrovei. J. Agric. Food Chem.
glycerol. Bioresour. https://doi.org/ 10.1021/jf070058y.
Technol. https://doi.org/10.1016/j.biortech.2013.05.030. Fisch, K.M., Bakeer, W., Yakasai, A.A., Song, Z., Pedrick, J., Wasil, Z., Bailey, A.M.,
Chang, G.F., Gao, N.S., Tian, G.W., Wu, Q.H., Chang, M., Wang, X.G., 2013b. Lazarus, C.M., Simpson, T.J., Cox, R.J., 2011. Rational domain swaps decipher
Improvement of docosahexaenoic acid production on glycerol by programming in fungal highly reducing polyketide synthases and resurrect an
Schizochytrium sp. S31 with constantly high oxygen transfer coefficient. extinct metabolite. J. Am. Chem. Soc. https://doi.org/10.1021/ja206914q.
Bioresour. Technol. https:// doi.org/10.1016/j.biortech.2013.04.107. Ganuza, E., Benítez, S.T., Atalah, Vega, O.O., Ganga, R., Izquierdo, M.S., 2008.
Chang, G.F., Wu, J., Jiang, C.H., Tian, G.W., Wu, Q.H., Chang, M., Wang, X.G., Crypthecodinium cohnii and Schizochytrium sp. as potential substitutes to
2014. The relationship of oxygen uptake rate and kLa with rheological fisheries- derived oils from seabream (Sparus aurata) microdiets. Aquaculture.
properties in high cell density cultivation of docosahexaenoic acid by https://doi. org/10.1016/j.aquaculture.2008.02.005.
Schizochytrium sp. S31. Bioresour. Gładkowski, W., Kiełbowicz, G., Chojnacka, A., Bobak, Ł., Spychaj, R., Dobrzanski,
Technol. https://doi.org/10.1016/j.biortech.2013.11.002. Z., ´ Trziszka, T., Wawrzenczyk, C., 2014. The effect of feed supplementation
Chang, K.J.L., Rye, L., Dunstan, G.A., Grant, T., Koutoulis, A., Nichols, P.D., with dietary ´ sources of n-3 polyunsaturated fatty acids, flaxseed and algae
Blackburn, S. I., 2015b. Life cycle assessment: heterotrophic cultivation of Schizochytrium sp., on their incorporation into lipid fractions of Japanese
thraustochytrids for biodiesel production. J. Appl. Phycol. quail eggs. Int. J. Food Sci.
https://doi.org/10.1007/s10811-014-0364-9. Technol. https://doi.org/10.1111/ijfs.12497.
Chang, M., Jing, G., Wang, X., Xiang, Y., Liu, Y., Rui, J., 2015a. A strategy for the Goldstein, S., Belsky, M., 1964. Avenic culture studies of a new marine
highly efficient production of docosahexaenoic acid by Aurantiochytrium Phycomycete possessing an unusual type of asexual reproduction. Am. J.
limacinum SR21 using glucose and glycerol as the mixed carbon sources. Bot. https://doi.org/ 10.1002/j.1537-2197.1964.tb06602.x.
Bioresour. Technol. https:// doi.org/10.1016/j.biortech.2014.11.046. Guo, D.S., Ren, L.J., Yin, F.W., Huang, H., Li, G.L., 2016. Development of a real-
Chen, C.Y., Yang, Y.T., 2018. Combining engineering strategies and fermentation time bioprocess monitoring method for docosahexaenoic acid production
technology to enhance docosahexaenoic acid (DHA) production from an by Schizochytrium sp. Bioresour. Technol. https://doi.org/10.1016/j.
indigenous Thraustochytrium sp. BM2 strain. Biochem. Eng. J. biortech.2016.05.044.
https://doi.org/10.1016/j. bej.2018.02.010. Guo, D.S., Ji, X.J., Ren, L.J., Li, G.L., Huang, H., 2017. Improving docosahexaenoic
Chen, C.Y., Lee, M.H., Dong, C.D., Leong, Y.K., Chang, J.S., 2020. Enhanced acid production by Schizochytrium sp. using a newly designed high-oxygen-
production of microalgal lipids using a heterotrophic marine microalga supply bioreactor. AICHE J. https://doi.org/10.1002/aic.15783.
Thraustochytrium sp. Guo, D.S., Ji, X.J., Ren, L.J., Li, G.L., Sun, X.M., Chen, K.Q., Gao, S., Huang, H.,
BM2. Biochem. Eng. J. https://doi.org/10.1016/j.bej.2019.107429. 2018. Development of a scale-up strategy for fermentative production of
Chen, W., Zhou, P., Zhu, Y., Xie, C., Ma, L., Wang, X., Bao, Z., Yu, L., 2016a. docosahexaenoic acid by Schizochytrium sp. Chem. Eng. Sci.
Improvement in the docosahexaenoic acid production of Schizochytrium https://doi.org/10.1016/j.
sp. S056 by replacement of sea salt. Bioprocess Biosyst. Eng. ces.2017.11.021.
https://doi.org/10.1007/s00449- 015-1517-1. Guo, D.S., Tong, L.L., Ji, X.J., Ren, L.J., Ding, Q.Q., 2020. Development of a
Chen, W., Zhou, P.P., Zhang, M., Zhu, Y.M., Wang, X.P., Luo, X.A., Bao, Z.D., Yu, strategy to improve the stability of culture environment for
L.J., 2016b. Transcriptome analysis reveals that up-regulation of the fatty acid docosahexaenoic acid fermentation by Schizochytrium sp. Appl. Biochem.
synthase gene promotes the accumulation of docosahexaenoic acid in Biotechnol. https://doi.org/10.1007/s12010- 020-03298-7.
Schizochytrium sp. S056 when glycerol is used. Algal Res. Gupta, A., Abraham, R.E., Barrow, C.J., Puri, M., 2015. Omega-3 fatty acid
https://doi.org/10.1016/j.algal.2016.02.007. Cheng, R., Ma, R., Li, K., Rong, H., production from enzyme saccharified hemp hydrolysate using a novel
Lin, X., Wang, Z., Yang, S., Ma, Y., 2012. marine Thraustochytrid strain. Bioresour. Technol.
Agrobacterium tumefaciens mediated transformation of marine microalgae https://doi.org/10.1016/j.biortech.2014.11.031.
Schizochytrium. Microbiol. Res. Han, X., Zhao, Z.N., Wen, Y., Chen, Z., 2020. Enhancement of docosahexaenoic
https://doi.org/10.1016/j.micres.2011.05.003. acid production by overexpression of ATP-citrate lyase and acetyl-CoA
Chi, Z., Pyle, D., Wen, Z., Frear, C., Chen, S., 2007. A laboratory study of carboxylase in Schizochytrium sp. Biotechnol. Biofuels.
producing docosahexaenoic acid from biodiesel-waste glycerol by https://doi.org/10.1186/s13068-020- 01767-z.
microalgal fermentation. Harris, J., Viner, K., Champagne, P., Jessop, P.G., 2018. Advances in microalgal
Process Biochem. https://doi.org/10.1016/j.procbio.2007.08.008. lipid extraction for biofuel production: a review: microalgal lipid extraction
Chodchoey, K., Verduyn, C., 2012. Growth, fatty acid profile in major lipid classes for biofuel production. Biofuels Bioprod. Biorefin.
and lipid fluidity of Aurantiochytrium Mangrovei Sk-02 as a function of https://doi.org/10.1002/bbb.1923.
growth temperature. Braz. J. Microbiol. https://doi.org/10.1590/S1517- Hauvermale, A., Kuner, J., Rosenzweig, B., Guerra, D., Diltz, S., Metz, J.G., 2006.
838220120001000020. Fatty acid production in Schizochytrium sp.: involvement of a
Chu, F.F., Cheng, J., Li, K., Wang, Y.G., Li, X., Yang, W.J., 2020. Enhanced lipid polyunsaturated fatty acid synthase and a type I fatty acid synthase. Lipids.
accumulation through a regulated metabolic pathway of phosphorus luxury https://doi.org/10.1007/s11745- 006-5025-6.
uptake in the microalga Chlorella vulgaris under nitrogen starvation and Hayashi, S., Satoh, Y., Ujihara, T., Takata, Y., Dairi, T., 2016. Enhanced production
phosphorus repletion. ACS Sustain. Chem. Eng. https://doi.org/10.1021/ of polyunsaturated fatty acids by enzyme engineering of tandem acyl
acssuschemeng.9b07447. carrier proteins. Sci. Rep-UK. https://doi.org/10.1038/srep35441.
Cui, G., Wang, Z., Hong, W., Liu, Y.J., Chen, Z., Cui, Q., Song, X., 2019. Enhancing Hayashi, S., Satoh, Y., Ogasawara, Y., Maruyama, C., Hamano, Y., Ujihara, T., Dairi,
tricarboxylate transportation-related NADPH generation to improve T., 2019. Control mechanism for cis double-bond formation by
biodiesel production by Aurantiochytrium. Algal Res. polyunsaturated fatty- acid synthases. Angew. Chem. Int. Ed.
https://doi.org/10.1016/j. https://doi.org/10.1002/anie.201812623.
algal.2019.101505. Hoang, C., Nguyen, S., Chia, H., Yu, Y.K., Dinh, T., 2018. Sugarcane bagasse as a
Cui, G.Z., Ma, Z., Liu, Y.J., Feng, Y., Sun, Z., Cheng, Y., Song, X., Cui, Q., 2016. novel carbon source for heterotrophic cultivation of oleaginous microalga
Overexpression of glucose-6-phosphate dehydrogenase enhanced the Schizochytrium sp. Ind. Crop. Prod.
polyunsaturated fatty acid composition of Aurantiochytrium sp. SD116. https://doi.org/10.1016/j.indcrop.2018.05.005.
Algal Res. https://doi.org/10.1016/j.algal.2016.08.005. Hoang, M.H., Nguyen, C., Pham, H.Q., Van, N.L., Hoang, D.L., Van, S.L., Hai, T.N.,
Dairi, T., Hayashi, S., Naka, M., Ikeuchi, K., Ohtsuka, M., Kobayashi, K., Satoh, Y., Ha, C. H., Nhan, L.D., Anh, H.T.L., 2016. Transcriptome sequencing and
Ogasawara, Y., Maruyama, C., Hamano, Y., 2019. Control mechanism for comparative analysis of Schizochytrium mangrovei PQ6 at different

14
F. Du et al. Biotechnology Advances 48 (2021) 107725
cultivation times. Biotechnol. Lett. https://doi.org/10.1007/s10529-016- Li, Z., Meng, T., Ling, X.P., Li, J., He, N., 2018b. Overexpression of malonyl-CoA:
2165-5. Acp transacylase in Schizochytrium sp. to improve polyunsaturated fatty
Hong, W.K., Heo, S.Y., Park, H.M., Kim, C.H., Sohn, J.H., Kondo, A., Seo, J.W., acids production. J. Agric. Food Chem.
2013a. https://doi.org/10.1021/acs.jafc.8b01026.
Characterization of a squalene synthase from the thraustochytrid microalga Li, Z., Ling, X., Zhou, H., Meng, T., Zeng, J., Hang, W., Shi, Y., He, N., 2019b.
Aurantiochytrium sp. KRS101. J. Microbiol. Biotechnol. Screening chemical modulators of benzoic acid derivatives to improve lipid
https://doi.org/10.4014/ jmb.1212.12023. accumulation in Schizochytrium limacinum SR21 with metabolomics
Hong, W.K., Yu, A., Oh, B.R., Park, J.M., Kim, C.H., Sohn, J.H., Kondo, A., Seo, J.W., analysis. Biotechnol. Biofuels. https://doi.org/10.1186/s13068-019-1552-2.
2013b. Large-scale production of microalgal lipids containing high levels of Lin, H.X., Lee, Y.K., 2017. Genetic engineering of medium-chain-length fatty acid
docosahexaenoic acid upon fermentation of Aurantiochytrium sp. KRS101. synthesis in Dunaliella tertiolecta for improved biodiesel production. J.
Food Nutr. Appl.
Res. https://doi.org/10.4236/fns.2013.49A1001. Phycol. https://doi.org/10.1007/s10811-017-1210-7.
Hsin-Ju, Hsieh, Su, Chia-Hung, Chien, Liang-Jung, 2012. Accumulation of lipid Liang, F., Lindblad, P., 2017. Synechocystis PCC 6803 overexpressing RuBisCO
production in Chlorella minutissima by triacylglycerol biosynthesis-related grow faster with increased photosynthesis. Metab. Eng. Commun.
genes cloned from Saccharomyces cerevisiae and Yarrowia lipolytica. J. https://doi.org/ 10.1016/j.meteno.2017.02.002.
Microbiol. https:// doi.org/10.1007/s12275-012-2041-5. Liang, Y.N., Sarkany, N., Cui, Y., Yesuf, J., Trushenski, J., Blackburn, J.W., 2010. Use
Huang, J., Aki, T., Yokochi, T., Nakahara, T., Honda, D., Kawamoto, S., Shigeta, S., of sweet sorghum juice for lipid production by Schizochytrium limacinum
Ono, K., Suzuki, O., 2003. Grouping newly isolated docosahexaenoic acid- SR21.
producing thraustochytrids based on their polyunsaturated fatty acid Bioresour. Technol. https://doi.org/10.1016/j.biortech.2009.12.087.
profiles and comparative analysis of 18S rRNA genes. Mar. Biotechnol. 5, Lippmeier, J.C., Crawford, K.S., Owen, C.B., Rivas, A.A., Metz, J.G., Apt, K.E.,
450–457. https://doi.org/10.1007/ s10126-002-0110-1. 2009. Characterization of both polyunsaturated fatty acid biosynthetic
Huang, J., Jiang, X., Zhang, X., Chen, W., Hu, S., 2008. Expressed sequence tag pathways in Schizochytrium sp. Lipids. https://doi.org/10.1007/s11745-009-
analysis of marine fungus Schizochytrium producing docosahexaenoic acid. 3311-9.
J. Biotechnol. https://doi.org/10.1016/j.jbiotec.2008.07.1994. Liu, T., Sanchez, J.F., Chiang, Y.M., Oakley, B.R., Wang, C.C.C., 2014a. Rational
Huang, T.Y., Lu, W.C., Chu, I.M., 2012. A fermentation strategy for producing domain swaps reveal insights about chain length control by ketosynthase
docosahexaenoic acid in Aurantiochytrium limacinum SR21 and increasing domains in fungal nonreducing polyketide synthases. Org. Lett.
C22:6 proportions in total fatty acid. Bioresour. Technol. https://doi.org/10.1021/ol5003384.
https://doi.org/10.1016/j. Liu, Y., Tang, J., Li, J., Daroch, M., Cheng, J.J., 2014b. Efficient production of
biortech.2012.07.068. triacylglycerols rich in docosahexaenoic acid (DHA) by osmo-heterotrophic
Humhal, T., Kastanek, P., Jezkova, Z., Cadkova, A., Kohoutkova, J., Branyik, T., marine protists. Appl. Microbiol. Biotechnol.
2017. https://doi.org/10.1007/s00253-014-6032-9.
Use of saline waste water from demineralization of cheese whey for Liu, Z.X., You, S., Tang, B.P., Wang, B., Sheng, S., Wu, F.A., Wang, J., 2019. Inositol
cultivation of Schizochytrium limacinum PA-968 and Japonochytrium as a new enhancer for improving lipid production and accumulation in
marinum AN-4. Bioprocess Biosyst. Eng. https://doi.org/10.1007/s00449- Schizochytrium sp. SR21. Environ. Sci. Pollut. Res.
016-1707-5. https://doi.org/10.1007/s11356-019-06056-3.
Innis, S.M., 2008. Dietary omega 3 fatty acids and the developing brain. Brain Makri, A., Fakas, S., Aggelis, G., 2010. Metabolic activities of biotechnological
Res. https://doi.org/10.1016/j.brainres.2008.08.078. interest in Yarrowia lipolytica grown on glycerol in repeated batch cultures.
Irshad, A., Sharma, A., Daniell, H., Shashi, K., 2015. Altered lipid composition Bioresour. Technol. https://doi.org/10.1016/j.biortech.2009.11.024.
and enhanced lipid production in green microalga by introduction of Marchan, L.F., Lee Chang, K.J., Nichols, P.D., Polglase, J.L., Mitchell, W.J.,
brassica diacylglycerol acyltransferase 2. Plant Biotechnol. J. Gutierrez, T., 2017. Screening of new British Thraustochytrids isolates for
https://doi.org/10.1111/ pbi.12278. docosahexaenoic acid (DHA) production. J. Appl. Phycol.
Jakobsen, A.N., Aasen, I.M., Josefsen, K.D., Strøm, A.R., 2008. Accumulation of https://doi.org/10.1007/s10811-017-1149-8.
docosahexaenoic acid-rich lipid in thraustochytrid Aurantiochytrium sp. Meesapyodsuk, D., Qiu, X., 2016. Biosynthetic mechanism of very long chain
strain T66: effects of N and P starvation and O-2 limitation. Appl. Microbiol. polyunsaturated fatty acids in Thraustochytrium sp. 26185. J. Lipid Res.
Biotechnol. https:// doi.org/10.1007/s00253-008-1537-8. https://doi. org/10.1194/jlr.M070136.
Jiang, Y., Fan, K.W., Wong, R.D.Y., Chen, F., 2004. Fatty acid composition and Merkx, J.A., Rasmussen, H., Muise, D.M., Benjamin, J.J.R., Kottwitz, H., Tanner,
squalene content of the marine microalga Schizochytrium mangrovei. J. K., Milway, M.T., Purdue, L.M., Scaife, M.A., Armenta, R.E., Woodhall, D.L.,
Agric. Food Chem. https://doi.org/10.1021/jf035004c. 2018. Engineering xylose metabolism in thraustochytrid T18. Biotechnol.
Jon Meadus, W., Duff, P., Rolland, D., Lynn Aalhus, J., Uttaro, B., Russell Dugan, Biofuels. https:// doi.org/10.1186/s13068-018-1246-1.
M.E., 2011. Feeding docosahexaenoic acid to pigs reduces blood Miller, M.R., Nichols, P.D., Carter, C.G., 2007. Replacement of fish oil with
triglycerides and induces gene expression for fat oxidation. Can. J. Chem. thraustochytrid Schizochytrium sp. L oil in Atlantic salmon parr (Salmo salar
https://doi.org/10.4141/cjas2011- 055. L) diets. Comp. Biochem. Phys. Part A.
Kim, H.U., Huang, A.H.C., 2004. Plastid lysophosphatidyl acyltransferase is https://doi.org/10.1016/j.cbpa.2007.05.018.
essential for embryo development in arabidopsis. Plant Physiol. Moran, C.A., Morlacchini, M., Keegan, J.D., Rutz, F., Fusconi, G., 2020.
https://doi.org/10.1104/ pp.103.035832. Docosahexaenoic acid enrichment of layer hen tissues and eggs through
Lee Chang, K.J., Dumsday, G., Nichols, P.D., Dunstan, G.A., Blackburn, S.I., dietary supplementation with heterotrophically grown Aurantiochytrium
Koutoulis, A., 2013. High cell density cultivation of a novel limacinum. J. Appl. Poult. Res. https://doi.
Aurantiochytrium sp. strain TC 20 in a fed-batch system using glycerol to org/10.1016/j.japr.2019.10.002.
produce feedstock for biodiesel and omega-3 oils.
Moreno-P´erez, A.J., Sanchez-García, A., Salas, J.J., Garc´ ´es, R., Martínez-Force,
Appl. Microbiol. Biotechnol. https://doi.org/10.1007/s00253-013-4965-z.
E., 2011. Acyl-ACP thioesterases from macadamia (Macadamia tetraphylla)
Leong, H.Y., Su, C.A., Lee, B.S., Lan, C.W., Law, C.L., Chang, J.S., Show, P.L., 2019.
nuts: cloning, characterization and their impact on oil composition. Plant
Development of Aurantiochytrium limacinum SR21 cultivation using salt-
Physiol. Biochem.
rich waste feedstock for docosahexaenoic acid production and application
https://doi.org/10.1016/j.plaphy.2010.10.002.
of natural colourant in food product. Bioresour. Technol.
Nagano, N., Sakaguchi, K., Taoka, Y., Okita, Y., Honda, D., Ito, M., Hayashi, M.,
https://doi.org/10.1016/j.
2011. Detection of genes involved in fatty acid elongation and desaturation
biortech.2018.09.093.
in Thraustochytrid Marine Eukaryotes. J. Oleo Sci.
Leyland, Ben, Leu, Stefan, Boussiba, Sammy, 2017. Are Thraustochytrids algae?
https://doi.org/10.5650/jos.60.475.
Fungal. Biol. Rev. https://doi.org/10.1016/j.funbio.2017.07.006.
Papanikolaou, S., Aggelis, G., 2011. Lipids of oleaginous yeasts. Part I:
Li, J., Liu, R., Chang, G., Li, X., Chang, M., Liu, Y., Jin, Q., Wang, X., 2015. A
Biochemistry of single cell oil production. Eur. J. Lipid Sci. Technol. 113 (8),
strategy for the highly efficient production of docosahexaenoic acid by
1031–1051. https://doi. org/10.1002/ejlt.201100014.
Aurantiochytrium limacinum SR21 using glucose and glycerol as the mixed
Papanikolaou, S., Sarantou, S., Komaitis, M., Aggelis, G., 2010. Repression of
carbon sources. Bioresour. Technol.
reserve lipid turnover in Cunninghamella echinulata and Mortierella
https://doi.org/10.1016/j.biortech.2014.11.046.
isabellina cultivated in multiple-limited media. J. Appl. Microbiol.
Li, J., Zhou, H., Pan, X., Li, Z., Ling, X., 2019a. The role of fluconazole in the
https://doi.org/10.1111/j.1365- 2672.2004.02376.x.
regulation of fatty acid and unsaponifiable matter biosynthesis in
Parsons, J.B., Rock, C., 2013. Bacterial lipids: metabolism and membrane
Schizochytrium sp. MYA 1381.
homeostasis.
BMC Microbiol. https://doi.org/10.1186/s12866-019-1622-4.
Prog. Lipid Res. https://doi.org/10.1016/j.plipres.2013.02.002.
Li, Q., Chen, G.Q., Fan, K.W., Lu, F.P., Aki, T., Jiang, Y., 2009. Screening and
Patel, A., Rova, U., Christakopoulos, P., Matsakas, L., 2019. Simultaneous
characterization of squalene-producing Thraustochytrids from Hong Kong
production of DHA and squalene from Aurantiochytrium sp. grown on
mangroves. J. Agric. Food Chem. https://doi.org/10.1021/jf9003972.
forest biomass hydrolysates.
Li, Z., Chen, X., Li, J., Meng, T., Wang, L., Chen, Z., Shi, Y., Ling, X., Luo, W., Liang,
Biotechnol. Biofuels. https://doi.org/10.1186/s13068-019-1593-6.
D., Lu, Y., Li, Q., He, N., 2018a. Functions of PKS genes in lipid synthesis of
Pereira, S.L., Huang, Y.S., Bobik, E.G., Kinney, A.J., Stecca, K.L., Packer, J.C.L.,
Schizochytrium sp. by gene disruption and metabolomics analysis. Mar.
Mukerji, P., 2004. A novel omega 3-fatty acid desaturase involved in the
Biotechnol. https://doi.org/10.1007/s10126-018-9849-x.

15
F. Du et al. Biotechnology Advances 48 (2021) 107725
biosynthesis of eicosapentaenoic acid. Biochem. J. enhance lipid production and prevent lipid peroxidation in Schizochytrium
https://doi.org/10.1042/BJ20031319. sp. Biotechnol.
Pour, A.K., Mirmehrabi, M., Brar, S.K., 2020. A novel process for isolation and Biofuels. https://doi.org/10.1186/s13068-018-1065-4.
purification of polyunsaturated fatty acids from a thraustochytrid. Algal Res. Takanori, M., Keishi, S., Rie, H., Takumi, K., Eriko, A., Yoichiro, H., Masahiro, H.,
https:// doi.org/10.1016/j.algal.2020.101806. Daiske, H., Yuji, O., Shinichi, S., Nozomu, O., Makoto, Ito, 2012. Analysis of
Pyle, D.J., Garcia, R.A., Wen, Z., 2008. Producing docosahexaenoic acid (DHA)- 12-fatty acid desaturase function revealed that two distinct pathways are
rich algae from biodiesel-derived crude glycerol: effects of impurities on active for the synthesis of PUFAs in T. aureum ATCC 34304. J. Lipid Res.
DHA production and algal biomass composition. J. Agric. Food Chem. https://doi.org/10.1016/ j.jpainsymman.2014.08.012.
https://doi.org/10.1021/ jf800602s. Tang, S.K., Qin, C.R., Wang, H.G., Li, S.F., Tian, S.J., 2011. Study on supercritical
Qi, F., Zhang, M., Chen, Y., Jiang, X., Lin, J., Cao, X., Huang, J., 2017. A extraction of lipids and enrichment of DHA from oil-rich microalgae. J.
lignocellulosic hydrolysate-tolerant Aurantiochytrium sp. mutant strain for Supercrit.
docosahexaenoic acid production. Bioresour. Technol. Fluids. https://doi.org/10.1016/j.supflu.2011.01.010.
https://doi.org/10.1016/j.biortech.2016.12.011. Varela, J., Pereira, H., Vila, M., Leon, R., 2015. Production of carotenoids by
Qiao, K., Wasylenko, T.M., Zhou, K., Xu, P., Stephanopoulos, G., 2017. Lipid microalgae: ´ achievements and challenges. Photosynth. Res.
production in Yarrowia lipolytica is maximized by engineering cytosolic https://doi.org/10.1007/s11120- 015-0183-0.
redox metabolism. Nat. Biotechnol. https://doi.org/10.1038/nbt.3763. Villarroel Hipp, M.P., Silva Rodriguez, D., 2018. Bioremediation of piggery
Qu, L., Ren, L.J., Huang, H., 2013. Scale-up of docosahexaenoic acid production slaughterhouse wastewater using the marine protist, Thraustochytrium
in fed- batch fermentation by Schizochytrium sp. based on volumetric kinney VAL- B1. J. Adv. Res. https://doi.org/10.1016/j.jare.2018.01.010.
oxygen-transfer coefficient. Biochem. Eng. J. Wang, F., Bi, Y., Diao, J., Lv, M., Zhang, W., 2019. Metabolic engineering to
https://doi.org/10.1016/j.bej.2013.05.011. enhance biosynthesis of both docosahexaenoic acid and odd-chain fatty
Ratledge, C., 2012. Omega-3 biotechnology: errors and omissions. Biotechnol. acids in
Adv. https://doi.org/10.1016/j.biotechadv.2012.04.002. Schizochytrium sp. S31. Biotechnol. Biofuels.
Ren, L.J., Zhuang, X.Y., Chen, S.L., Ji, X.J., Huang, H., 2015. Introduction of ω-3 https://doi.org/10.1186/s13068-019- 1484-x.
desaturase obviously changed the fatty acid profile and sterol content of Wang, K., Sun, T., Cui, J., Liu, L., Bi, Y., Pei, G., Chen, L., Zhang, W., 2018a.
Schizochytrium sp. J. Agric. Food Chem. Screening of chemical modulators for lipid accumulation in Schizochytrium
https://doi.org/10.1021/acs.jafc.5b04238. sp S31. Bioresour.
Ren, L.J., Sun, G.N., Ji, X.J., Hu, X.C., Huang, H., 2014. Compositional shift in lipid Technol. https://doi.org/10.1016/j.biortech.2018.03.104.
fractions during lipid accumulation and turnover in Schizochytrium sp. Wang, Q., Sen, B., Liu, X., He, Y., Xie, Y., Wang, G., 2018b. Enhanced saturated
Bioresour. Technol. https://doi.org/10.1016/j.biortech.2014.01.078. fatty acids accumulation in cultures of newly-isolated strains of
Ren, L.J., Sun, X.M., Ji, X.J., Chen, S.L., Guo, D.S., Huang, H., 2017. Enhancement Schizochytrium sp. and Thraustochytriidae sp. for large-scale biodiesel
of docosahexaenoic acid synthesis by manipulation of antioxidant capacity production. Sci. Total Environ.
and prevention of oxidative damage in Schizochytrium sp. Bioresour. https://doi.org/10.1016/j.scitotenv.2018.03.078.
Technol. https:// doi.org/10.1016/j.biortech.2016.10.040. Wang, S., Lan, C., Wang, Z., Wan, W., Song, X., 2020. Optimizing
Ren, L.J., Chen, S.L., Geng, L.J., Ji, X.J., Xu, X., Song, P., Gao, S., Huang, H., 2018. eicosapentaenoic acid production by grafting a heterologous polyketide
Exploring the function of acyltransferase and domain replacement in order synthase pathway in the Thraustochytrid Aurantiochytrium. J. Agric. Food
to change the polyunsaturated fatty acid profile of Schizochytrium sp. Algal Chem. https://doi.org/10.1021/acs. jafc.0c04299.
Res. https://doi. org/10.1016/j.algal.2017.11.021. Wang, Z.X., Jian, Z.G., Fang, H., Prior, B.A., 2001. Glycerol production by
Ruiz-Rosa, I., Garcia-Rodriguez, F.J., Mendoza-Jimenez, J., 2016. Development microbial fermentation: a review. Biotechnol. Adv.
and application of a cost management model for wastewater treatment https://doi.org/10.1016/S0734-9750(01) 00060-X.
and reuse processes. J. Clean. Prod. Xie, X., Meesapyodsuk, D., Qiu, X., 2017. Ketoacylsynthase domains of a PUFA
https://doi.org/10.1016/j.jclepro.2015.12.044. synthase in Thraustochytrium can effectively function as stand-alone
Ryu, B.G., Kim, K., Kim, J., Han, J.I., Yang, J.W., 2013. Use of organic waste from enzymes in Escherichia coli. Appl. Environ. Microbiol.
the brewery industry for high-density cultivation of the docosahexaenoic https://doi.org/10.1128/AEM.03133-16.
acid-rich microalga, Aurantiochytrium sp. KRS101. Bioresour. Technol. Xin, F., Wu, Y.R., He, J., 2014. Simultaneous fermentation of glucose and xylose
https://doi.org/ 10.1016/j.biortech.2012.11.049. to butanol by Clostridium sp. strain BOH3. Appl. Environ. Microbiol.
Sakaguchi, K., Matsuda, T., Kobayashi, T., Ohara, J., Hamaguchi, R., Abe, E., https://doi.org/ 10.1128/AEM.00337-14.
Nagano, N., Hayashi, M., Ueda, M., Honda, D., Okita, Y., Taoka, Y., Sugimoto, Yamasaki, T., Aki, T., Shinozaki, M., Taguchi, M., Kawamoto, S., Ono, K., 2006.
S., Okino, N., Ito, M., 2012. Versatile transformation system that is Utilization of Shochu distillery wastewater for production of
applicable to both multiple transgene expression and gene targeting for polyunsaturated fatty acids and xanthophylls using Thraustochytrid. J.
Thraustochytrids. Appl. Environ. Biosci. Bioeng. https://doi.org/ 10.1016/j.enzmictec.2017.11.009.
Microbiol. https://doi.org/10.1128/AEM.07129-11. Yan, J., Cheng, R., Lin, X., You, S., Ma, Y., 2012. Overexpression of acetyl-CoA
Schellenberg, G.D., Sarthy, A., Larson, A.E., Backer, M.P., Furlong, C.E., 1984. synthetase increased the biomass and fatty acid proportion in microalga
Xylose isomerase from Escherichia coli. Characterization of the protein and Schizochytrium. Appl. Microbiol. Biotechnol. 97 (5), 1933–1939.
the structural gene. J. Biol. Chem. https://doi.org/10.1007/978-3-540- https://doi.org/10.1007/s00253-012- 4481-6.
85387-9_1. Ye, J.R., Liu, M.M., He, M.X., Ye, Y., Huang, J.C., 2019. Illustrating and enhancing
Schmidt, I., Schewe, H., Gassel, S., Jin, C., Buckingham, J., Hümbelin, M., the biosynthesis of astaxanthin and docosahexaenoic acid in
Sandmann, G., Schrader, J., 2011. Biotechnological production of Aurantiochytrium sp. SK4.
astaxanthin with Phaffia rhodozyma/Xanthophyllomyces dendrorhous. Mar. Drugs. https://doi.org/10.3390/md17010045.
Appl. Microbiol. Biotechnol. https://doi. org/10.1007/s00253-010-2976-6. Ye, H.K., He, Y.D., Xie, Y.X., Sen, B., Wang, G.Y., 2020. Fed-batch fermentation of
Shirasaka, N., Hirai, Y., Nakabayashi, H., Yoshizumi, H., 2005. Effect of mixed carbon source significantly enhances the production of
cyanocobalamin and p-toluic acid on the fatty acid composition of docosahexaenoic acid in Thraustochytriidae sp PKU#Mn16 by differentially
Schizochytrium limacinum (Thraustochytriaceae, Labyrinthulomycota). regulating fatty acids biosynthetic pathways. Bioresour. Technol.
Mycoscience. https://doi.org/10.1007/ s10267-005-0259-3. https://doi.org/10.1016/j.
Singh, D., Mathur, A.S., Tuli, D.K., Puri, M., Barrow, C.J., 2015. Propyl gallate and biortech.2019.122402.
butylated hydroxytoluene influence the accumulation of saturated fatty Yin, F.W., Guo, D.S., Ren, L.J., Ji, X.J., Huang, H., 2018a. Development of a
acids, omega-3 fatty acid and carotenoids in Thraustochytrids. J. Funct. method for the valorization of fermentation wastewater and algal-residue
Foods. https://doi. org/10.1016/j.jff.2015.03.022. extract in docosahexaenoic acid production by Schizochytrium sp.
Song, X., Ma, Z., Tan, Y., Zhang, H., Cui, Q., 2017. Wastewater recycling Bioresour. Technol. https:// doi.org/10.1016/j.biortech.2018.06.109.
technology for fermentation in polyunsaturated fatty acid production. Yin, F.W., Zhang, Y.T., Jiang, J.Y., Guo, D.S., Gao, S., Gao, Z., 2018b. Efficient
Bioresour. Technol. https:// doi.org/10.1016/j.biortech.2017.03.034. docosahexaenoic acid production by Schizochytrium sp. via a two-phase pH
Suen, Y.L., Tang, H., Huang, J., Chen, F., 2014. Enhanced production of fatty acids control strategy using ammonia and citric acid as pH regulators. Process
and astaxanthin in Aurantiochytrium sp. by the expression of Vitreoscilla Biochem. https:// doi.org/10.1016/j.procbio.2018.11.013.
hemoglobin. J. Agric. Food Chem. https://doi.org/10.1021/jf5048578. Yokoyama, R., Honda, D., 2007. Taxonomic rearrangement of the genus
Sun, H., Chen, H., Zang, X., Hou, P., Zhou, B., Liu, Y., Wu, F., Cao, X., Zhang, X., Schizochytrium sensu lato based on morphology, chemotaxonomic
2015. Application of the Cre/loxP site-specific recombination system for characteristics, and 18S rRNA gene phylogeny (Thraustochytriaceae,
gene transformation in Aurantiochytrium limacinum. Molecules. Labyrinthulomycetes): emendation for Schizochytrium and erection of
https://doi.org/10.3390/ molecules200610110. Aurantiochytrium and Oblongichytrium gen. nov.
Sun, L.N., Ren, L.J., Zhuang, X.Y., Ji, X.J., Yan, J.C., Huang, H., 2014. Differential Mycoscience. https://doi.org/10.1007/s10267-006-0362-0.
effects of nutrient limitations on biochemical constituents and Yu, X.J., Sun, J., Zheng, J.Y., Sun, Y.Q., Wang, Z., 2016. Metabolomics analysis
docosahexaenoic acid production of Schizochytrium sp. Bioresour. Technol. reveals 6- benzylaminopurine as a stimulator for improving lipid and DHA
https://doi.org/10.1016/j. accumulation of Aurantiochytrium sp. J. Chem. Technol. Biotechnol.
biortech.2014.02.106. https://doi.org/10.1002/ jctb.4869.
Sun, X.M., Ren, L.J., Bi, Z.Q., Ji, X.J., Zhao, Q.Y., Jiang, L., Huang, H., 2018. Zhang, A.Q., Xie, Y.X., He, Y.D., Wang, W.J., Sen, B., Wang, G.Y., 2019. Bio-based
Development of a cooperative two-factor adaptive-evolution method to squalene production by Aurantiochytrium sp. through optimization of

16
F. Du et al. Biotechnology Advances 48 (2021) 107725
culture conditions, and elucidation of the putative biosynthetic pathway
genes. Bioresour.
Technol. https://doi.org/10.1016/j.biortech.2019.121415.
Zhang, K., Chen, L., Liu, J., Gao, F., He, R., Chen, W., Guo, W., Chen, S., Li, D.,
2017. Effects of butanol on high value product production in
Schizochytrium limacinum B4D1. Enzym. Microb. Technol.
https://doi.org/10.1016/j.enzmictec.2017.03.007.
Zhang, L., Zhao, H., Lai, Y., Wu, J., Chen, H., 2013. Improving docosahexaenoic
acid productivity of Schizochytrium sp by a two-stage AEMR/shake mixed
culture mode.
Bioresour. Technol. https://doi.org/10.1016/j.biortech.2013.05.072.
Zhang, Y., Ward, V., Dennis, D., Plechkova, N.V., Armenta, R., Rehmann, L., 2018.
Efficient extraction of a docosahexaenoic acid (DHA)-rich lipid fraction from
Thraustochytrium sp. using lonic liquids. Mater. https://doi.org/10.3390/
ma11101986.
Zhao, X., Ren, L., Guo, D., Wu, W., Ji, X., Huang, H., 2016. CFD investigation of
Schizochytrium sp impeller configurations on cell growth and
docosahexaenoic acid synthesis. Bioprocess Biosyst. Eng.
https://doi.org/10.1007/s00449-016-1608-7.
Zheng, H.L., Gao, Z., Yin, F.W., Ji, X.J., Huang, H., 2012. Lipid production of
Chlorella vulgaris from lipid-extracted microalgal biomass residues through
two-step enzymatic hydrolysis. Bioresour. Technol.
https://doi.org/10.1016/j. biortech.2012.04.007.

17

You might also like