Xie 2021

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Progress in Lipid Research 82 (2021) 101095

Contents lists available at ScienceDirect

Progress in Lipid Research


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

Review

Regulation of long-chain polyunsaturated fatty acid biosynthesis in


teleost fish
Dizhi Xie a, 1, Cuiying Chen b, 1, Yewei Dong c, 1, Cuihong You c, Shuqi Wang b, *, Óscar Monroig d, *,
Douglas R. Tocher b, e, Yuanyou Li a, *
a
College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
b
Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
c
Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510642, China
d
Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (IATS-CSIC), 12595 Castellón, Spain
e
Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling FK94LA, Scotland, United Kingdom

A R T I C L E I N F O A B S T R A C T

Keywords: Omega-3 (n-3) long-chain polyunsaturated fatty acids (LC-PUFA, C20-24), including eicosapentaenoic acid (EPA,
LC-PUFA biosynthesis 20:5n-3) and docosahexaenoic acid (DHA, 22:6n-3), are involved in numerous biological processes and have a
Transcriptional regulation range of health benefits. Fish have long been considered as the main source of n-3 LC-PUFA in human diets.
Post-transcriptional regulation
However, the capacity for endogenous biosynthesis of LC-PUFA from C18 PUFA varies in fish species based on the
Aquaculture
presence, expression and activity of key enzymes including fatty acyl desaturases (Fads) and elongation of very
long-chain fatty acids (Elovl) proteins. In this article, we review progress on the identified Fads and Elovl, as well
as the regulatory mechanisms of LC-PUFA biosynthesis both at transcriptional and post-transcriptional levels in
teleosts. The most comprehensive advances have been obtained in rabbitfish Siganus canaliculatus, a marine
teleost demonstrated to have the entire pathway for LC-PUFA biosynthesis, including the roles of transcription
factors hepatocyte nuclear factor 4α (Hnf4α), liver X receptor alpha (Lxrα), sterol regulatory element-binding
protein 1 (Srebp-1), peroxisome proliferator-activated receptor gamma (Pparγ) and stimulatory protein 1
(Sp1), as well as post-transcriptional regulation by individual microRNA (miRNA) or clusters. This research has,
for the first time, demonstrated the involvement of Hnf4α, Pparγ and miRNA in the regulation of LC-PUFA
biosynthesis in vertebrates. The present review provides readers with a relatively comprehensive overview of
the progress made into understanding LC-PUFA biosynthetic systems in teleosts, and some insights into
improving endogenous LC-PUFA biosynthesis capacity aimed at reducing the dependence of aquafeeds on fish oil
while maintaining or increasing flesh LC-PUFA content and the nutritional quality of farmed fish.

1. Introduction basket providing, not only high-quality protein, but also n-3 (or “omega-
3”) long-chain (C20-24) polyunsaturated fatty acids (n-3 LC-PUFA) such
Fish and seafood are important components of the human food as eicosapentaenoic acid (EPA, 20:5n-3) and docosahexaenoic acid

Abbreviations: ALA, α-linolenic acid (18:3n-3); ARA, arachidonic acid (20:4n-6); PUFA, polyunsaturated fatty acid; C/EBP, CCAAT/enhancer binding protein;
DBD, DNA binding domain; DHA, docosahexaenoic acid (22:6n-3); DPA, docosapentaenoic acid (22:5n-3); DR-1, direct repeat 1; EFA, essential fatty acid; Elovl,
elongation of very long-chain fatty acids; EMSA, electrophoretic mobility shift assay; EPA, eicosapentaenoic acid (20:5n-3); FA, fatty acid; Fads, fatty acyl desaturase;
FO, fish oil; Hnf4α, hepatocyte nuclear factor 4α; LA, linoleic acid (18:2n-6); LBD, ligand binding domain; LC-PUFA, long-chain (C20-24) polyunsaturated fatty acid;
lncRNA, long non-coding RNA; Lxr, liver X receptor; LRE, LXR response element; miRNA, microRNA; MUFA, monounsaturated fatty acid; ncRNA, non-coding RNA;
NF-1, nuclear factor 1; NF-Y, nuclear factor Y; Ppar, peroxisome proliferator activated receptor; PPRE, peroxisome proliferator response element; PUFA, poly­
unsaturated fatty acid (≥ 2 double bonds); RXR, retinoid X receptor; SCHL, Siganus canaliculatus hepatocyte line; siRNA, small interfering RNA; Sp1, stimulatory
protein 1; SRE, sterol response element; Srebp-1, sterol-regulatory element binding protein 1; TF, transcription factor; TSS, transcription start site; UTR, untranslated
region; VLC-PUFA, very long-chain (>C24) polyunsaturated fatty acid; VO, vegetable oil.
* Corresponding authors.
E-mail addresses: sqw@stu.edu.cn (S. Wang), oscar.monroig@csic.es (Ó. Monroig), yyli16@scau.edu.cn (Y. Li).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.plipres.2021.101095
Received 6 November 2020; Received in revised form 24 February 2021; Accepted 12 March 2021
Available online 16 March 2021
0163-7827/© 2021 Published by Elsevier Ltd.
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

(DHA, 22:6n-3). n-3 LC-PUFA are physiologically important compounds and regulation of the biosynthetic pathways. This will provide readers
required for normal growth and development of vertebrates including with a comprehensive understanding of the state-of-the-art of LC-PUFA
humans, and can play key roles in mitigating inflammatory, cardiovas­ biosynthesis in teleost fish. The research reviewed in this article is key to
cular and cerebrovascular diseases, and are beneficial in some types of gain insight into the capacity that farmed fish have to utilize dietary oil
cancer [1,2]. Seafood in general and oily fish in particular are unique sources currently used in aquafeed formulations to satisfy both their
sources of n-3 LC-PUFA for humans, unlike terrestrial animals and their own physiological demands and produce food with high nutritional
products (e.g. milk and eggs) in which these essential nutrients are ab­ value (i.e. n-3 LC-PUFA rich) for humans.
sent or at very low levels. While annual global fish production has
increased from 19 million tons in 1950 to 179 million tons in 2018 [3], 2. Pathways of LC-PUFA biosynthesis in teleost fish
capture fisheries production has remained stagnant for some decades,
while provision of seafood from aquaculture has been steadily Fish, like all vertebrates, cannot synthesize LA or ALA de novo due to
increasing and currently accounts for almost 53 % of total production the lack of Δ12 and Δ15 desaturases, respectively, enzymes found only
[3], with prospects to reach up to 60–70 % by the year 2030 [4]. in plants, marine microbes including microalgae, heterotrophic protists
In addition to being important for human health, n-3 LC-PUFA are and bacteria, and invertebrates [14–16]. Therefore, LA and ALA are
also essential nutrients ensuring normal growth and development of fish essential dietary nutrients for all vertebrates. The biosynthesis of LC-
themselves. The presence of n-3 LC-PUFA in fish derives both from their PUFA from C18 precursors requires a series of desaturation and elon­
diet and endogenous production (biosynthesis), with the latter varying gation reactions catalyzed by fatty acyl desaturase (Fads)2 enzymes and
substantially with species [5]. Historically, it has been widely accepted elongation of very long-chain fatty acid (Elovl) proteins (Fig. 1). As
that freshwater and salmonid species have significant LC-PUFA described below, several members of the Fads and Elovl protein families
biosynthetic capacity and so they can convert the C18 polyunsaturated participate in these pathways. It is largely accepted that LC-PUFA
fatty acids (PUFA), α-linolenic acid (ALA, 18:3n-3) and linoleic acid (LA, biosynthesis from LA and ALA is usually initiated by a ∆6 desatura­
18:2n-6), to the biologically active LC-PUFA including the n-3 EPA and tion, followed by an elongation and subsequent ∆5 desaturation leading
DHA, and the n-6 arachidonic acid (ARA, 20:4n-6). In contrast, marine to the production of ARA and EPA, respectively (Fig. 1). An alternative
teleosts were believed to have generally very limited or no capacity to pathway, termed the “Δ8 pathway”, starts with a chain elongation,
biosynthesize LC-PUFA from C18 PUFA precursors. The dichotomy of followed by Δ8 and then Δ5 desaturations to similarly enable ARA and
freshwater/salmonid fish vs. marine fish having high and low LC-PUFA EPA production from LA and ALA, respectively [24,25].
biosynthetic capacity, respectively, is now perceived as too simplistic Two distinct pathways allow vertebrates to biosynthesize DHA from
according to recent discoveries on the repertoire and function of key EPA. Arguably, the more widespread pathway for DHA biosynthesis
enzymes involved in these pathways, as well as the mechanisms regu­ among vertebrates is the so-called “Sprecher pathway”, consisting of
lating their activity. While this will be covered in detail in the present two consecutive elongations of EPA to produce 24:5n-3, followed by a
review, it is important to note that, regardless of the species’ habitat, ∆6 desaturation to form 24:6n-3, which then undergoes partial
understanding the ability of farmed fish for LC-PUFA biosynthesis has β-oxidation to DHA [18]. This pathway was first described in rats [26]
practical implications for fish health and wellbeing. and rainbow trout Oncorhynchus mykiss [27], and subsequently
For fish species with high LC-PUFA biosynthesizing capacity, phys­ described in other teleosts [12,28,29]. Alternatively, DHA can be also
iological demand for essential fatty acid (EFA) can be met through di­ synthesized through a more direct pathway termed the “∆4 pathway” by
etary supply of the C18 biosynthetic precursors LA and ALA present in which docosapentaenoic acid (DPA, 22:5n-3) is directly ∆4 desaturated
vegetable oils (VO) used widely in aquafeeds. However, endogenous to DHA (Fig. 1). This pathway was first reported in vertebrates in the
production of LC-PUFA from dietary precursors typically leads to lower marine teleost Siganus canaliculatus [19], and was subsequently shown
levels of n-3 LC-PUFA in the fillet and, to compensate lower nutritional to be present in several other teleosts (Table 1) [12,30–33].
and commercial value, fish fed VO-rich diets during most of the pro­
duction cycle can be fed a “finishing diet” with high inclusion of fish oil
(FO) before harvest [6]. On the contrary, for species with low capacity of 2.1. Fads enzymes
LC-PUFA biosynthesis, supply of pre-formed physiologically important
LC-PUFA is required, which is achieved practically with inclusion of FO Fads enzymes introduce unsaturation (double bonds) between a pre-
rich in LC-PUFA and, to a lesser extent, fishmeal in feed formulations existing double bond and the carboxyl end of the fatty acyl chain, and
[7]. Algal products, particularly microalgal oils, can contain high con­ thus they are also termed “front-end” desaturases. Compared to mam­
tents of n-3 LC-PUFA and thus have potential to replace FO in aquafeeds mals, which possess two distinct Fads-like desaturases including FADS1
[7]. Additionally, owing to rapidly changing environmental factors (e.g. (∆5 desaturase) and FADS2 (∆6 desaturase) [34], teleosts have a rather
temperature and salinity), fish species with LC-PUFA biosynthesis ability different fads gene complement (Fig. 1) [20]. With the exception of the
also have specific requirements for n-3 LC-PUFA for maintaining mem­ Elopomorpha species like Japanese eel Anguilla japonica, which has both
brane functionality and cellular osmoregulation [8–10]. The finite na­ Fads1 and Fads2 desaturases [20], virtually all teleosts possess only
ture of FO as a resource and its high price associated with an increasing Fads2 as the sole Fads-like desaturase in their genomes [5,13]. However,
demand to satisfy a growing industry has resulted in VO being unlike mammals, teleosts can have a varied number of fads2 genes, such
increasingly used in aquafeeds. As mentioned above, such a strategy as one (e.g. zebrafish Danio rerio), two (e.g. rabbitfish S. canaliculatus),
represents a metabolic challenge for species with low LC-PUFA biosyn­ three (e.g. Nile tilapia Oreochromis niloticus), or even four (e.g. Atlantic
thetic capacities and this has prompted considerable interest in under­ salmon Salmo salar) genes, while other species such as the pufferfish
standing the regulatory mechanisms involved in LC-PUFA biosynthesis
of fish, especially farmed species, so as to develop farming strategies that
2
enable efficient and effective utilization of dietary VO [7,10–12]. Gene/protein nomenclature: In this review, the gene/protein symbols used
The present paper reviews the substantial progress made recently on for human, mouse and teleosts follow the nomenclature guidelines of human
(Homo sapiens), mouse (Mus musculus) and zebrafish (Danio rerio), respectively.
the regulatory mechanisms of LC-PUFA biosynthesis at both transcrip­
Using as example “Fads2”, the human gene is referred to as “FADS2” and its
tional and post-transcriptional levels in teleosts. As the pathways of LC-
protein as “FADS2”; mouse genes are also italicized but lower case other than
PUFA biosynthesis in vertebrates including fish have been reviewed in the first letter “Fads2”, while proteins are the same as for human proteins
detail recently [5,13], these are only summarized to provide the “FADS2”; for zebrafish and other fish, genes are entirely lower case and itali­
appropriate context while the focus of the review is on providing readers cized "fads2", while proteins are also lower case other than the first letter and
with knowledge of the molecular mechanisms underpinning the control not italicized "Fads2".

2
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

Fig. 1. Biosynthetic pathways of LC-PUFA in teleosts. General characteristics of LC-PUFA biosynthetic capability and the key enzymatic genes in teleosts are as
follow: 1) freshwater fish, salmonids and a few marine teleosts such as rabbitfish Siganus canaliculatus have LC-PUFA biosynthetic capacity, whereas most marine
teleosts such as Atlantic cod Gadus morhua, grouper Epinephelus coioides and golden pompano Trachinotus ovatus either lack or have only weak capability due to the
absence of Δ5 Fads2 activity [5,13,17]; 2) There are two distinct pathways of DHA biosynthesis from DPA: the widespread “Sprecher pathway” [18], and the “∆4
pathway” that exists in a few teleosts such as S. canaliculatus [19]; 3) Several Fads types were found in teleosts, e.g. both ∆5 Fads1 and ∆6 Fads2 in Japanese eel
Anguilla japonica [20], three ∆6 Fads2 and one ∆5 Fads2 in Atlantic salmon (Salmo salar) [12,21,22], three Fads2 (∆6∆5 Fads2, ∆4 Fads2, and one unidentified Fads2)
in tilapia (Oreochromis niloticus) [12], two Fads2 (∆6∆5 Fads2 and ∆4 Fads2) in rabbitfish S. canaliculatus [19], and one ∆6∆5 Fads2 in zebrafish (Danio rerio) [23].
“∆x” denotes reactions catalyzed by fatty acyl desaturases, Fads; “Elo” indicates reactions catalyzed by elongation of very long-chain fatty acid proteins, Elovl.

Table 1
Fatty acyl desaturases identified in teleosts.
Fads type Habitat Fish species References

Δ6 Fads2 Freshwater fish Common carp Cyprinus carpio; Eurasian perch Perca fluviatilis; grass carp Ctenopharyngodon idellus; mandarin fish Siniperca [35–38]
chuatsi; pirarucu Arapaima gigas; rainbow trout Oncorhynchus mykiss
Diadromous fish Atlantic salmon Salmo salar; Japanese eel Anguilla japonica; meagre Argyrosomus regius; pike eel Muraenesox cinereus [21,39–41]
Marine fish Atlantic bluefin tuna Thunnus thynnus; Atlantic cod Gadus morhua; Asian sea bass/barramundi Lates calcarifer; black [35,42–54]
seabream Acanthopagrus schlegelii; Chu’s croaker Nibea coibor; cobia Rachycentron canadum; gilthead seabream Sparus
aurata; grouper Epinephelus coioides; Japanese flounder Paralichthys olivaceu; Nibe croaker Nibea mitsukurii; salema Sarpa
salpa; blackspotted croaker Nibea diacanthus; European sea bass Dicentrarchus labrax; spotted scat Scatophagus Argus; thick-lip
grey mullet Chelon labrosus; turbot Psetta maxima
Amphidromous Boddart’s goggle-eyed goby Boleophthalmus boddarti [55]
fish
Δ5 Fads2 Freshwater fish O. mykiss [56]
Diadromous fish A. japonica (Δ5 Fads1) [20]
Marine fish C. labrosus; Senegalese sole Solea senegalensis; S. salpa [30,33]
Δ6Δ5 Freshwater fish African catfish Clarias gariepinus; Mexican silverside Chirostoma estor; silver barb Barbonymus gonionotus; striped snakehead [12,23,31,57–61]
Fads2 Channa striata; tambaqui Colossoma macropomum; tench Tinca tinca; Nile tilapia Oreochromis niloticus; zebrafish Danio rerio
Diadromous fish S. salar [22]
Marine fish Rabbitfish Siganus canaliculatus; sand sole Pegusa lascaris [19,33]
Δ4 Fads2 Freshwater fish C. estor; C. striata; medaka Oryzias latipes; O. niloticus [12,31,32]
Marine fish Golden pompano Trachinotus ovatus; P. lascaris; S. canaliculatus; S. senegalensis; sand smelt Atherina presbyter [19,30,42,62]

Takifugu rubripes and Tetraodon nigroviridis have no fads genes [5,13]. Since its first report in S. canaliculatus [19], the ∆4 Fads2 has been
Likely compensating for the lack of fads1 in their genomes, teleost found in several other fish species (Table 1) [12,30–33], which may
Fads2 have functionally diversified during evolution [13] and a sum­ indicate it is more widespread in teleosts than initially believed. Inter­
mary of the Fads desaturase activities reported in freshwater, diadro­ estingly, Δ8 desaturation activity has been reported for almost all the
mous and marine teleost species can be found in Table 1. Some fish presently analyzed Fads2, which indicates that Δ8 desaturation is likely
species, such as zebrafish [23], African catfish (Clarias gariepinus) [57] an intrinsic enzymatic ability within vertebrate Fads2 [25]. A general
and silver barb (Barbonymus gonionotus) [58], possess one fads2 with conclusion from the above studies is that carnivorous marine fish have a
bifunctional Δ6Δ5 desaturation activity. Other species including limited LC-PUFA biosynthetic capacity due to having only a single fads2
rainbow trout have two distinct fads2 with separate Δ5 and Δ6 desa­ with Δ6 desaturase activity and, thus, they lack the ability for the Δ5
turation activities [35,56]. Other teleosts including rabbitfish [19], pike desaturation required for biosynthesis of EPA and ARA. The acquisition
silverside (Chirostoma estor) [31] and striped snakehead (Channa striata) of several copies of fads2 with functional diversification has enabled an
[32,61] also have two fads2 genes that were characterized as coding for enhanced capacity for endogenous production of DHA in some teleosts.
enzymes with Δ6Δ5 and Δ4 desaturase activities. Three copies of fads2 Thus, a recent study demonstrated that this process enabled some
genes have been isolated from Nile tilapia, with two encoding Δ6Δ5 sticklebacks to successfully colonize habitats that are naturally depleted
bifunctional and Δ4 monofunctional desaturases, respectively, and one in DHA [63]. Furthermore, it was also shown that the acquisition of
with unknown function [12]. Furthermore, four fads2 genes have been various DHA biosynthetic pathways enabled flatfishes to colonize
found in Atlantic salmon, with three exhibiting Δ6 desaturase activity freshwater environments [64].
and one being essentially a Δ5 desaturase [12,21,22]. The varying
numbers of fads2 genes have likely arisen as a result of both whole
genome duplication and tandem duplication events that have occurred 2.2. Elongation of very long-chain fatty acids (Elovl) proteins
in teleosts, which provided “spare” genetic material for the adaptive
mutation of fads2-type genes, and the regaining of Δ5 activity in some Elovl catalyze the usually rate-limiting step in the multi-enzyme
species [5,13]. complex enabling the two-carbon elongation of pre-existing fatty acyl
chains. Seven members of the ELOVL family (ELOVL1-7) have been

3
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

identified in mammals based on their protein motif sequences and Table 2


substrate specificity [34,65]. Generally, ELOVL1, ELOVL3, ELOVL6 and Elongation of very long-chain fatty acids (Elovl) proteins identified in teleosts.
ELOVL7 have the ability to elongate saturated fatty acids (SFA) and Elovl Habitat Species References
monounsaturated fatty acids (MUFA), while the preferred substrates of type
ELOVL2, ELOVL4 and ELOVL5 are PUFA [65–68]. Since the first report Elovl5 Freshwater fish African catfish [32,36,58–60,69–74]
of the molecular and functional characterization of an Elovl5 in a teleost, Clarias gariepinus;
zebrafish [69], many studies have reported the characterization of PUFA Eurasian perch
elongases including Elovl2, Elovl4 and Elovl5, from a large variety of Perca fluviatilis;
grass carp
fish species (see [5,13]). Table 2 provides a list of teleost species in Ctenopharyngodon
which at least one PUFA Elovl has been functionally characterized. idellus; mandarin
Briefly, Elovl5 is present in virtually all teleosts, and is responsible for fish Siniperca
the elongation of primarily C18 and C20 PUFA substrates (e.g., chuatsi; Northern
pike Esox Lucius;
[32,41,52,75]). Some Elovl5 from euryhaline herbivorous species
rainbow trout
including rabbitfish, spotted scat (Scatophagus argus) [85], salema (Sarpa Oncorhynchus
salpa) [33], and the carnivorous fish, Chinese perch (Siniperca chuatsi) mykiss; striped
[33] and sand sole (Pegusa lascaris) [33], have been reported to also have snakehead Channa
some elongation capacity towards C22 PUFA. Elovl2 has a preference for striata; tambaqui
Colossoma
C20 and C22 PUFA, with some residual capability to elongate C18 PUFA macropomum; silver
[13], and has been identified in freshwater zebrafish [94], African cat­ barb Barbonymus
fish [57], tench (Tinca tinca) [60], salmonids S. salar [75] and O. mykiss gonionotus; tench
[74], and the catadromous Japanese eel [95]. The expansion of teleosts Tinca tinca; Nile
tilapia Oreochromis
during evolution has been accompanied by a loss of the elovl2 gene in the
niloticus; zebrafish
more recently emerged lineages that include the majority of marine Danio rerio
species [13]. Such loss of elongation capacity has been hypothesized to Diadromous Atlantic salmon [39,41,75,76]
further explain the limited capability of marine fish for LC-PUFA fish Salmo salar;
biosynthesis alluded to above. While this argument is still valid for Japanese eel
Anguilla japonica;
most marine fish, including many farmed species, a recent study re­
meagre
ported the presence of a functional elovl2 in the marine European Argyrosomus regius;
sardine (Sardina pilchardus) [96]. Along with Elovl5 and Elovl2, Elovl4 pike eel Muraenesox
has been widely studied in fish, with data confirming that teleosts cinereus
Marine fish Asian sea bass Lates [30,33,42,49,50,52,70,77–85]
generally have two distinct elovl4 genes, elovl4a and elovl4b, with
calcarifer; Atlantic
varying elongation capacities toward C18-22 PUFA substrates from which bluefin tuna
they can produce the so-called very long-chain (>C24) PUFA (VLC- Thunnus thynnus;
PUFA), with chains up to 36 carbons [29,69,81,83–87,90,92]. Impor­ Atlantic cod Gadus
tantly, beyond their role in VLC-PUFA biosynthesis, Elovl4 enzymes morhua; black
seabream
have a role in the biosynthesis of LC-PUFA such as DHA since they can
Acanthopagrus
elongate EPA and DPA to 24:5n-3, a key intermediate of the Sprecher schlegeli; Chu’s
pathway. Such elongation ability of teleost Elovl4 enzyme has been croaker Nibea
hypothesized to compensate for the absence of Elovl2 in many marine coibor; cobia
Rachycentron
teleost species alluded to above [86]. A recently uncovered elongase
canadum; gilthead
with putative roles in LC-PUFA biosynthesis is elovl8, for which two seabream Sparus
genes, elovl8a and elovl8b, exist in teleosts [97]. This novel elongase, aurata; golden
apparently absent in mammals [97], has been shown to have elongation pompano
activity toward C18 and C20 PUFA in teleosts, as demonstrated by Trachinotus ovatus;
Japanese flounder
functional assays of Elovl8 enzymes of African catfish [71] and rabbit­
Paralichthys
fish [97]. olivaceus; large
Although generally not involved in LC-PUFA biosynthesis pathways, yellow croaker
the other members of the Elovl family have also been studied in teleosts. Larimichthys crocea;
Nibe croaker Nibea
Zebrafish possess two elovl1 genes, elovl1a and elovl1b, and knockdown
mitsukurii; orange-
of elovl1 increased levels of C14 to C20 SFA, MUFA and PUFA in zebrafish spotted grouper
embryos, suggesting Elovl1 in zebrafish may also be involved in the Epinephelus coioides;
elongation of PUFA [98], different from human ELOVL1 that has high rabbitfish Siganus
activity toward C20 and C22 SFA and MUFA [99]. However, molecular canaliculatus;
salema Sarpa salpa;
characterization and nutritional regulation of Elovl6 was reported in
sand sole Pegusa
loach (Misgurnus anguillicaudatus) [100], large yellow croaker (Lar­ lascaris;
imichthys crocea) [101] and rainbow trout [102], with loach Elovl6 blackspotted
shown to elongate 16:0 and 16:1 to 18:0 and 18:1, respectively, similar croaker Nibea
to mammals [100]. While Elovl3 and Elovl7 have not yet been reported diacanthus;
Senegalese sole Solea
in fish, compared to mammalian Elovl7, Elovl7 from the marine crus­ senegalensis; spotted
tacean orange mud crab (Scylla olivacea) had elongation activity towards scat Scatophagus
16:1n-7 and, to a lower extend, C18 PUFA [103]. argus; turbot Psetta
maxima; southern
bluefin tuna
3. The transcriptional regulation of LC-PUFA biosynthesis in Thunnus maccoyii;
teleosts thick-lip grey
(continued on next page)
Based on the reported activities of their Fads and PUFA Elovl

4
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

Table 2 (continued ) Table 3


Elovl Habitat Species References
Teleost species that have been reported to have a complete LC-PUFA biosyn­
type thetic pathway established by either possessing at least Fads with Δ6 and Δ5
activities and elongation capacity toward C18-22 PUFA or, alternatively, Fads
mullet Chelon
with Δ6, Δ5 and Δ4 activities and C18-20 elongation capacity.
labrosus
Amphidromous Boddart’s goggle- [55] Teleosts Fads and Elovl References
fish eyed goby
Freshwater species
Boleophthalmus
African catfish Clarias Δ6/Δ5 Fads2, Elovl5, Elovl4, [57,71]
boddarti
gariepinus Elovl2, Elovl8
Elovl4 Freshwater fish C. gariepinus; [71,86,87]
Mexican silverside Δ6Δ5 Fads2, Δ4 Fads2, Elovl5 [12,31]
D. rerio; loach
Chirostoma estor
Misgurnus
Rainbow trout Oncorhynchus Δ6 Fads2, Δ5 Fads2, Elovl5, [35,56,70,74]
anguillicaudatus;
mykiss Elovl4, Elovl2
O. mykiss
Silver barb Barbonymus Δ6Δ5 Fads2, Elovl5 [58]
Diadromous S. salar [88]
gonionotus
fish
Striped snakehead Channa Δ6Δ5 Fads2, Δ4 Fads2, Elovl5 [32,61]
Marine fish A. schlegeli; [29,50,81,83,85,89–93]
striata
E. coioides;
Tambaqui Colossoma Δ6Δ5 Fads2, Elovl5 [59]
L. crocea; Nibea
macropomum
mitsukurii;
Tench Tinca tinca Δ6Δ5 Fads2, Elovl5, Elovl2 [60]
R. canadum;
Tilapia Oreochromis niloticus Δ6Δ5 Fads2, Δ4 Fads, Elovl5 [12,70]
S. aurata; S.
Zebrafish Danio rerio Δ6Δ5 Fads2, Elovl5, Elovl4, [23,69,86,94]
canaliculatus;
Elovl2
S. argus;
S. senegalensis; Diadromous species
T. ovatus; T. thynnus Atlantic salmon Salmo salar Δ6 Fads2, Δ6Δ5 Fads2, Elovl5, [12,21,22,75,88]
Elovl2 Freshwater fish C. gariepinus; [57,59,60,74,94] Elovl4, Elovl2
C. macropomum; Japanese eel Anguilla Δ6 Fads2, Δ5 Fads1, Elovl5, [20,41,95]
D. rerio; O. mykiss; japonica Elovl2
T. tinca
Marine species
Diadromous A. japonica; Salmo [75,95]
fish solar Rabbitfish Siganus Δ6Δ5 Fads2, Δ4 Fads2, Elovl5, [19,83,97]
Marine fish Sardina pilchardus [96] canaliculatus Elovl4, Elovl8
Elovl8 Freshwater fish C. gariepinus [70,97] Salema Sarpa salpa Δ6 Fads2, Δ5 Fads2, Elovl5 [33,51]
Marine fish S. canaliculatus [97] Sand sole Pegusa lascaris Δ6Δ5 Fads2, Δ4 Fads2, Elovl5 [33,51]
Thicklip grey mullets Chelon Δ6 Fads2, Δ5 Fads2, Elovl5 [33,51]
labrosus

enzymes (Tables 1 and 2), and considering that Δ6 and Δ5 desaturation


capacity, as well as C18-22 PUFA elongation capacity, are required for stimulatory protein 1 (SP1) were also found in the Δ6 FADS2 promoter
biosynthesis of LC-PUFA (ARA, EPA and DHA) from the C18 biosynthetic [106]. Activation of PPARα was shown to promote Δ6 FADS2 promoter
precursors LA and ALA, a total of 15 fish species have been reported to activity [106] and the expression of Elovl5 [105]. In addition, PPARγ
have complete LC-PUFA biosynthetic pathways (Table 3). These include agonist troglitazone significantly decreased Δ6 FADS2 mRNA level and
nine freshwater, two diadromous and four marine fish. In addition, the enzymatic activity, which suggested that PPARγ would potentially
ability of fish to biosynthesize LC-PUFA depends, not only on their down-regulate LC-PUFA biosynthesis [107]. Other studies showed
complement of desaturase and elongase genes and their corresponding human Δ6 FADS2 and mouse Elovl5 genes were not direct targets of liver
enzyme activities, but also on the levels of gene expression at both X receptor α (LXRα), but were indirectly activated through the effects of
transcriptional and post-transcriptional levels. Thus, understanding the LXRα on SREBP-1c gene by combining with the LXR response element
regulatory mechanisms of key enzymatic gene expression may be (LRE) on the latter promoter [104,105]. Moreover, the expression of Δ6
helpful for developing methods and strategies to increase the ability for FADS2, Δ5 FADS1 and ELOVL5 were significantly increased by agonists
endogenous LC-PUFA biosynthesis in farmed fish species. of LXRα, which indicted that an LXRα–SREBP-1c pathway plays a reg­
It is well known that transcription is a vital step in the central dogma ulatory role in LC-PUFA biosynthesis in mammals [108]. It is only more
of transition from genes contained in chromosomes to active proteins. recently that similar studies have been performed in fish and here we
Transcription factors (TF) or “trans-acting elements” with specific acti­ summarize the major progress made in understanding the transcrip­
vator or repressor roles bind to cis-acting elements with corresponding tional regulation of LC-PUFA biosynthesis in teleosts.
nucleotide sequences in the promoter region of target genes, regulating
the initiation efficiency of RNA polymerase and gene expression. The
promoter region is defined as the upstream non-coding sequence relative 3.1. Cloning and characterization of fads2 and elovl gene promoters
to the first base-pair of the first exon of the target gene, also called the
transcription start site (TSS), and with position set as “+1” for promoter In order to investigate the transcriptional regulation of LC-PUFA
analysis purposes. Thus, the investigation of transcriptional regulation biosynthesis in teleosts, the upstream sequences from the initiation
of LC-PUFA biosynthesis usually follows the following main steps: (1) codon ATG of fads2 and elovl genes were cloned as the candidate pro­
cloning and characterization of fads2 and elovl promoters; (2) identifi­ moters and structures characterized. To date, the promoter of Δ6 fads2
cation of the key TF regulating the expression of fads2 and elovl target has been cloned in Atlantic salmon [109], Atlantic cod (Gadus morhua)
genes; (3) confirmation of the transcription regulatory effects and [109], European seabass (Dicentrarchus labrax) [110], Japanese seabass
mechanisms of TF in LC-PUFA biosynthesis. (Lateolabrax japonicus) [111], rainbow trout [112], large yellow croaker
In the last two decades, research into the transcriptional regulation [112], and orange-spotted grouper (Epinephelus coioides) [113]. More­
of mammal LC-PUFA biosynthesis was mainly focused on human Δ6 over, the promoter of Δ6Δ5 fads2 has been cloned in rabbitfish [114]
FADS2 [104] and mouse Elovl5 genes [105]. Binding sites for the TF and zebrafish [115], and Δ4 fads2 promoter has been cloned from rab­
sterol-regulatory element binding protein-1c (SREBP-1c) were identified bitfish [116] and golden pompano (Trachinotus ovatus) [62]. Regarding
in the promoters of Δ6 FADS2 [104] and Elovl5 genes [105], while those elovl, the promoter of elovl5 has been cloned in Atlantic salmon [117],
for peroxisome proliferator activated receptor α (PPARα) and large yellow croaker [81], orange-spotted grouper [82], golden pom­
pano [80], rabbitfish [118], zebrafish and snakehead [119], while the

5
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

elovl4 promoter has been cloned from large yellow croaker [112] and and includes three isoforms (SREBP-1a, -1c and -2), and SREBP-1c was
golden pompano [93]. shown to bind SRE in gene promoters and regulate target gene expres­
After the upstream sequences of the target genes were cloned, the sion [127]. LXR, including LXRα and LXRβ, belong to the nuclear hor­
core promoter regions were identified through progressive deletion mone receptor superfamily, which can heterodimerize with retinoid X
mutation and analysis by dual luciferase reporter assays. Binding sites receptor (RXR) and bind to the LXR response element (LRE) of SREBP-1c
for TF were identified in the promoters of target genes by bioinformatic promoter [128]. As mentioned above, an LXRα–SREBP-1c pathway
analysis, site-directed mutation and functional assays. In teleosts, mul­ plays a regulatory role in mammalian LC-PUFA biosynthesis
tiple TF binding sites were confirmed to be present in the core promoters [104,105,108]. In teleosts, srebp-1 and lxrα cDNAs have been isolated
of teleost fads2 and elovl. Sp1 binding element was identified in the from Atlantic salmon [129,130], rainbow trout [129], Japanese seabass
promoter of Atlantic salmon Δ6 fads2 [109], rabbitfish Δ6Δ5 fads2 and [131] and rabbitfish [132]. As to the regulatory roles of Srebp1 and Lxr
elovl5 [116,118], and C/EBP (CCAAT/enhancer binding protein) in LC-PUFA biosynthesis in teleosts, Lxrα was suggested to be a potential
element was identified in the Atlantic cod Δ6 fads2 promoter [109]. NF- up-regulator of elovl5 [130], and the Lxrα-Srebp-1 pathway was reported
1 (nuclear factor 1) element was found in the promoter of rabbitfish Δ4 to up-regulate the transcription of Δ6 fads2, elovl5a and elovl5b in
fads2 and Δ6Δ5 fads2 [114,116], and PPRE (peroxisome proliferator Atlantic salmon [133]. This pattern of regulation was also confirmed in
response element, binding site for PPAR) was discovered in the promoter rabbitfish [132] and large yellow croaker [81]. Recently, the presence of
of rabbitfish Δ6Δ5 fads2 [116], and golden pompano Δ4 fads2, elovl5 SRE in the core regulatory region of the elovl5 promoter of snakehead
and elovl4a [62,80,93]. DR-1 (direct repeat 1, binding site for hepato­ and zebrafish was demonstrated, and transcriptional activation of elovl5
cyte nuclear factor 4α, HNF4α) element was reported in the promoter of expression by Srebp-1 was confirmed [119]. These studies confirmed
rabbitfish Δ4 fads2, Δ6Δ5 fads2 and elovl5 [114,116,118]. The most that the Lxrα-Srebp-1 pathway is conserved between teleost and
conserved cis-acting elements, NF-Y (nuclear factor Y) and SRE (sterol mammals.
response element, binding site for SREBP), were confirmed in almost all
the reported fads2 and elovl gene promoters, including those of the 3.2.3. Pparα and Pparγ
Atlantic salmon Δ6 fads2 and elovl5 [109,117], Atlantic cod Δ6 fads2 PPAR, a family of ligand-activated nuclear hormone receptors with
[109], rabbitfish Δ6Δ5 fads2, Δ4 fads2 and elovl5 [114,116,118], DBD and LBD domains, includes three subtypes, namely PPARα, PPARβ
zebrafish Δ6Δ5 fads2 and elovl5 [115,119], and snakehead elovl5 [119]. and PPARγ [134]. Similar to LXR, activated PPAR heterodimerize with
RXR, and typically bind to DNA elements containing a common
3.2. Identification of TF regulating the expression of fads2 and elovl genes consensus response element that consists of two copies of the binding
site –AGGTCA– separated by n nucleotides (DRn) [135]. As mentioned
As described above, the structural analysis of promoters of fads2 and above, PPARα up-regulated mammalian LC-PUFA biosynthesis by pro­
elovl genes from teleosts uncovered a series of functional cis elements, moting the expression of Elovl5 and Δ6 FADS2 [105,106], while PPARγ
such as DR-1, SRE, NF-Y, Sp1 and PPRE [114,116,118], which may play has a potential down-regulation effect [107]. In teleosts, the regulation
vital roles in promoter activity and, thus, are likely to be involved in the of LC-PUFA biosynthesis by Pparα and Pparγ appeared more compli­
regulation of LC-PUFA biosynthesis in teleost fish. Subsequently, bind­ cated than in mammals. Pparα up-regulated fads2 expression and
ing/interaction of the TFs with the promoters was confirmed by elec­ enhanced LC-PUFA production in rainbow trout [112], and similar re­
trophoretic mobility shift assays (EMSA) and mass spectrometry sults were confirmed in a series of studies in golden pompano
[114–119]. Moreover, the roles of the corresponding TF, including [62,80,93]. However, the above study on rainbow trout also indicated
Hnf4α, Srebp-1, Lxr, Sp1 and Ppar, in the transcriptional regulation of that Pparα had no significant influence on fads2 expression in Japanese
LC-PUFA biosynthesis have been extensively investigated at two anal­ seabass and large yellow croaker [112]. Consistent with this, Pparα had
ysis levels including promoter activity and gene expression [120–122]. no effect on key enzymes involved in LC-PUFA biosynthesis in rabbitfish,
Promoter activity was investigated through dual luciferase reporter whereas Pparγ down-regulated expression of the Δ6Δ5 fads2 gene and
assay, and gene expression was examined by quantitative polymerase decreased LC-PUFA biosynthesis in rabbitfish [121].
chain reaction (qPCR). In addition, TF overexpression, agonist or in­
hibitor treatment, and small interfering RNA (siRNA) were also applied 3.2.4. Sp1
in the study of promoter activity and gene expression. The main TFs SP1, a ubiquitous trans-activator containing three contiguous
involved in the transcriptional regulation of LC-PUFA biosynthesis in carboxyl-terminal Cys2His2 zinc-fingers, is a basal TF maintaining the
teleosts and likely vertebrates in general are as follows: expression of housekeeping genes lacking a TATA-box [136]. The
binding sites for SP1 often contain GC-rich motifs in the promoters of
3.2.1. Hnf4α target genes [137]. Five predicted SP1 elements were found in the
Hnf4α is a major transcriptional regulator of lipid homeostasis. In human FADS2 promoter [106]. Reed et al. [138] inferred that SP1 may
mammals, HNF4α contains two typical structural domains, a DNA regulate mammal LC-PUFA biosynthesis by acting as a vital co-factor of
binding domain (DBD) and a ligand binding domain (LBD) [123]. In SREBP, because the SP1 binding sites are often very close to SRE [139].
teleosts, analysis of the hepatic transcriptome of Atlantic salmon showed Comparing the Δ6 fads2 promoter sequences of Atlantic salmon, Atlantic
that high expression of hepatic lipid transport-related genes was related cod, orange-spotted grouper, European seabass and Japanese seabass,
to alterations of hnf4α expression [124]. Additionally, the binding site of the Sp1 binding element was only detected in salmon and not in the
Hnf4α was predicted in the promoter of cptIα1b and cptIα2a genes in marine species [110,111], which suggested that the lack of Sp1 binding
grass carp (Ctenopharyngodon idella), which suggested that Hnf4α may element may account for the low promoter activities of Δ6 fads2 of those
be involved in modulating fatty acid β-oxidation [125]. In recent years, species and, possibly, in part explain the low LC-PUFA biosynthetic
our group discovered that Hnf4α was involved in the regulation of LC- capacity of marine carnivorous fish [109,110]. Consistent with this
PUFA biosynthesis in rabbitfish by targeting the promoters of Δ6Δ5 hypothesis, the insertion of the Sp1 binding element of rabbitfish Δ6Δ5
fads2, Δ4 fads2, and elovl5, and up-regulating the expression of these fads2 promoter into rabbitfish Δ4 fads2 or orange-spotted grouper Δ6
genes [114,120,126]. Indeed, these studies were the first to indicate that fads2 promoters significantly increased their promoter activities
Hnf4α was involved in the regulation of LC-PUFA biosynthesis in [113,118]. Subsequently, the Sp1 element was discovered in the elovl5
vertebrates. promoter of rabbitfish, snakehead and zebrafish [118,119], and Sp1 was
demonstrated to be involved in LC-PUFA biosynthesis by upregulating
3.2.2. Srebp-1 and Lxrα the expression of liver desaturase and elongase genes in rabbitfish [118],
SREBP belongs to the basic helix–loop–helix leucine zipper family and thus may be an important regulator of LC-PUFA biosynthetic genes

6
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

in teleosts. inhibited by Hnf4α inhibitors or siRNA against hnf4α mRNA [114,120].


Moreover, after injection of Hnf4α agonist, inhibitor or Hnf4α siRNA
into rabbitfish abdomen to modulate hnf4α expression, transcript levels
3.3. The transcriptional regulation of LC-PUFA biosynthesis in rabbitfish of Δ4 fads2 and Δ6Δ5 fads2 were positively correlated with the
expression level of the hnf4α gene [126]. The effect of Hnf4α regulation
The rabbitfish S. canaliculatus is a small commercially important on LC-PUFA biosynthesis of SCHL cells transfected with hnf4α mRNA or
marine teleost widespread along the Indo-West Pacific coast and one of liver of rabbitfish injected with Hnf4α agonist or inhibitor was deter­
the main species harvested (Fig. 2). In recent years, S. canaliculatus mined by analyzing fatty acid compositions of cellular and hepatic
farming has expanded rapidly in south-eastern Asia, including China, lipids. This showed that Hnf4α not only increased elovl5 promoter ac­
due to its popularity with consumers and with a market size of around tivity and elovl5 mRNA level, but also increased LC-PUFA biosynthesis
150 g. It usually feeds on macroalgae and seagrasses in nature, whereas both in vitro and in vivo [122]. These data demonstrated that Hnf4α
in farming it is often fed chopped fish but can also accept formula feed involved in the regulation of LC-PUFA biosynthesis in rabbitfish by
after simple training. As the first marine teleost demonstrated to have increasing transcription of Δ4 fads2, Δ6Δ5 fads2 and elovl5, which was
the capability for biosynthesis of LC-PUFA from C18 PUFA precursors the first demonstration of the role of Hnf4α in vertebrate LC-PUFA
[140,141] and have its complete PUFA biosynthesis pathway charac­ biosynthesis.
terized [19,25,83,97], the rabbitfish arose as a good model for the
investigation of LC-PUFA biosynthesis in teleosts. Consequently, rab­ 3.3.3. Positive roles of Lxrα-Srebp-1 in the transcriptional regulation of
bitfish is arguably the teleost species in which the transcriptional reg­ rabbitfish LC-PUFA biosynthesis
ulatory mechanisms of LC-PUFA biosynthesis have been most To investigate the role of the classical Lxrα-Srebp-1 pathway in the
comprehensively studied. In addition, the establishment of the regulation of LC-PUFA biosynthesis in rabbitfish, lxrα and srebp-1 genes
S. canaliculatus hepatocyte line (SCHL) provided an important experi­ were first cloned and characterized [132]. In vivo, the expression levels
mental tool [142]. Below summarizes the recent progress made into our of srebp-1 and the key LC-PUFA biosynthesis genes, Δ4 fads2, Δ6Δ5
understanding of the regulation of LC-PUFA biosynthesis in rabbitfish. fads2 and elovl5, displayed consistent changes in response to dietary
lipid source and ambient salinity [132]. Additionally, in vitro, the
3.3.1. Promoter characteristics of three key desaturase and elongase genes expression levels of lxrα and the key enzymatic genes were stimulated by
for rabbitfish LC-PUFA biosynthesis the Lxrα agonist T0901317 in rabbitfish primary hepatocytes. Besides,
In rabbitfish, the promoter sequences of three key LC-PUFA biosyn­ the promoter activities of Δ6Δ5 fads2 and elovl5 genes were significantly
thesis genes including Δ4 fads2, Δ6Δ5 fads2 and elovl5 have been cloned increased by over-expression of srebp-1 mRNA in HEK 293T cells, and
and characterized. In the promoter region of Δ4 fads2, several key ele­ the expression of Δ6Δ5 fads2 and elovl5 was decreased by knockdown of
ments including GATA-2, C/EBP, NF-1, NF-Y, SRE and DR-1 were pre­ srebp-1 mRNA in SCHL cells [143]. The above results demonstrated that
dicted, and the binding of Hnf4α and Nf-1 to the promoters was the Lxrα-Srebp-1 pathway is involved in LC-PUFA biosynthesis in rab­
confirmed using EMSA and LC-MS analyses [116]. In the core region of bitfish, as demonstrated previously in mammals [104,105,107,108],
the Δ6Δ5 fads2 promoter, elements such as C/EBP, NF-1, Sp1, NF-Y, through stimulating expression of Δ4 fads2, Δ6Δ5 fads2 and elovl5
SRE, AP1, DR-1 and PPRE were predicted, and the binding of Hnf4α genes.
and NF-1 were also confirmed [114]. Additionally, NF-Y, SRE and DR-1
elements were predicted in the elovl5 promoter, and interaction between 3.3.4. Role of Sp1 in determining the transcriptional activity of genes
Hnf4α and the DR-1 element was confirmed [122]. In summary, the core involved in the rabbitfish LC-PUFA biosynthesis
promoter regions of all three genes shared some similar structure, with An Sp1 binding site with high activity was found in rabbitfish Δ6Δ5
all being close to the TSS and containing binding sites for Hnf4α (DR-1) fads2 and elovl5 promoters, and overexpression of sp1 mRNA in SCHL
and Srebp-1c (NF-Y, SRE), while the promoters of both Δ6Δ5 fads2 and cells significantly increased the expression of Δ6Δ5 fads2 and elovl5, as
elovl5 genes contained the Sp1 element, and PPRE, the specific element well as the LC-PUFA content of the cells [118]. In contrast, the Sp1
for PPARγ, was found only in the Δ6Δ5 fads2 promoter. binding site was absent from the rabbitfish ∆4 fads2 promoter, but the
activity of this promoter was significantly increased by insertion of the
3.3.2. Stimulatory role of Hnf4α in the transcriptional regulation of Sp1 element from the rabbitfish ∆6∆5 fads2 promoter [118]. In addition,
rabbitfish LC-PUFA biosynthesis as described above, the activity of orange-spotted grouper fads2 pro­
The role of Hnf4α in the transcriptional regulation of rabbitfish LC- moter, which lacked an Sp1 binding site, was also significantly increased
PUFA biosynthesis has been demonstrated by both in vitro and in vivo after acquiring the Sp1 binding site of rabbitfish Δ6Δ5 fads2 [113].
experiments. Overexpression of Hnf4α in HEK 293T cells or hnf4α mRNA These results demonstrated that Sp1 is an important transcription factor
in SCHL cells, significantly increased Δ4 fads2 promoter activity and the involved in the regulation of LC-PUFA biosynthesis by targeting Δ6Δ5
expression level of Δ4 fads2 mRNA, which confirmed Hnf4α as an fads2 and elovl5 in rabbitfish [118], which was the first demonstration of
activator of the Δ4 fads2 gene [116]. In SCHL cells, the expression level this mechanism in a vertebrate.
of the Δ6Δ5 fads2 gene was also increased by Hnf4α agonists and
3.3.5. Inhibitory role of Pparγ in the transcriptional regulation of rabbitfish
LC-PUFA biosynthesis
In mammals, PPARγ is a key regulator involved in adipocyte differ­
entiation and lipid homeostasis [144] but, to the best of our knowledge,
there has been no report on the regulatory roles of PPARγ in mammalian
LC-PUFA biosynthesis. In rabbitfish primary hepatocytes, the Pparγ
agonists 2-bromopalmitate and fenofibrate increased the expression of
both pparγ and elovl5, but decreased the expression of Δ6Δ5 fads2 [145].
Moreover, the PPRE binding site for Pparγ was identified in the pro­
moter of Δ6Δ5 fads2 gene, and site-directed mutation and EMSA indi­
cated interaction between Pparγ and the promoter of the Δ6Δ5 fads2
gene [121]. Specifically, over-expression of pparγ in SCHL cells inhibited
the expression of Δ6Δ5 fads2 and reduced LC-PUFA biosynthesis ca­
Fig. 2. Rabbitfish Siganus canaliculatus. pacity, while pparγ RNAi knockdown increased the expression of Δ6Δ5

7
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

fads2. These data demonstrated a negative effect of Pparγ in the regu­ srebp-1, Δ6 fads2, elovl5 and elovl4 mRNA decreased with high dietary
lation of rabbitfish LC-PUFA biosynthesis [121]. LC-PUFA [81,112,146]. As these data suggested that there might be a
regulatory relationship between Lxrα, Srebp-1 and key LC-PUFA
3.4. Studies on the transcriptional regulation of LC-PUFA biosynthesis in biosynthesis genes in this species, the Δ6 fads2, elovl5 and elovl4
other teleosts cDNAs and their corresponding promoters were characterized
[81,112,146]. In HEK 293T cells, the over-expression of lxrα signifi­
Besides rabbitfish, other relevant studies on the transcriptional cantly increased the activities of the elovl4 and elovl5 promoters, while
regulation of LC-PUFA biosynthesis have been conducted in Atlantic over-expression of srebp-1 significantly decreased the promoter activ­
salmon, Japanese and European seabass, large yellow croaker, golden ities of Δ6 fads2 and elovl4 [81,112]. These results indicated that Lxrα
pompano, zebrafish and gilthead seabream (Sparus aurata). The main and Srebp-1 were involved in the transcriptional regulation of key genes
results are summarized below. in the LC-PUFA biosynthesis pathway. Additionally, in croaker primary
hepatocytes, expression of the elovl4 gene was up-regulated by an Lxrα
3.4.1. Atlantic salmon agonist and decreased by a Srebp-1 inhibitor, while elovl5 expression
Studies on transcriptional regulation of LC-PUFA biosynthesis in was increased by Lxrα agonist but showed no response to the Srebp-1
teleosts was initiated in Atlantic salmon which, second to rabbitfish, is inhibitor [112]. These results confirmed that the transcription of
the species with the most knowledge of the key enzymes of LC-PUFA elovl4 can be regulated by both Lxrα and Srebp-1, while suggesting that
biosynthesis. To date, genes including Δ6 fads2 (a/b/c), Δ5 fads2, the expression of elovl5 might be the target of Lxrα, but not Srebp-1
elovl5 (a/b), elovl2 and elovl4b have been cloned and functionally [112].
characterized from this species [12,21,22,35,70,73,75,88,117]. In
addition, mechanisms of transcriptional regulation of LC-PUFA biosyn­ 3.4.4. Golden pompano
thesis have also been investigated in Atlantic salmon. Specifically, the An in vivo feeding trial with golden pompano juveniles suggested that
salmon head kidney cell line was treated with EPA and DHA, and a this species could not synthesize LC-PUFA from C18 PUFA or that such
positive relationship between the relative expression of the transcription ability is very limited [147]. However, full-length mRNAs encoding two
factors lxr and srebp1 and expression levels of key desaturase and putative Elovl elongases and two putative Fads2 desaturases were iso­
elongase target genes (Δ6 fads2a, Δ5 fads2, elovl5a and elovl2) was lated, and the corresponding enzymatic activities were investigated by
observed [130], indicating a possible role for the Lxrα-Srebp1 pathway heterologous expression in yeast [62,80,93]. The two putative elongases
in the regulation of LC-PUFA biosynthesis in salmon. Later, the regula­ were reported to possess Elovl4 and Elovl5 activities, while both Fads2
tory role of Lxrα-Srebp1 was confirmed by TF ligand activation and were reported to display Δ8/Δ5/Δ4 desaturase activities [62,80,93].
agonist assays, as well as srebp1 over-expression, which established that Although no Δ6 activity was detected, these data would still suggest that
Lxr can activate Srebp1, and Srebp1 was an activator of Δ6 fads2, elovl5a golden pompano would have the ability to biosynthesize EPA and ARA
and elovl5b expression [133]. Moreover, the conserved elements NF-Y from ALA and LA, respectively, via the Δ8 pathway and, furthermore,
and SRE, as well as the Sp1 binding element, were found in the core also have the ability to produce DHA from EPA via the Δ4 pathway.
promoter of Atlantic salmon Δ6 fads2 and elovl5 genes through site- These data are unusual and highly unexpected as they are not consistent
directed mutation and EMSA assay [109,117]. with the data from the in vivo feeding trial in golden pompano [147] or
the existing body of knowledge on LC-PUFA biosynthesis and teleost
3.4.2. Japanese and European seabass phylogeny [5,13]. However, if and when the data are confirmed, they
In Japanese seabass, promoter activity analysis showed that over- suggest that pompano will represent another highly interesting species
expression of srebp-1 or pparα significantly increased the activity of for further study. In this respect, so far the promoters of elovl5 [80],
fads2 promoter, which confirmed the importance of NF-Y and SRE sites elovl4 [93], fads2 [62] have been isolated, and promoter activity assays
for fads2 gene expression in this species of seabass [112]. Studies in have shown that Pparαb was the key positive TF in the regulation of
seabass were also the first to report on the transcriptional regulation of these genes [62,80,93], similar to the regulatory mechanisms in rainbow
LC-PUFA biosynthesis at the level of epigenetic modification in teleosts. trout [112] and mammals [106].
Specifically, the influence of different dietary lipid sources on gene
expression and core promoter methylation of Δ6 fads2 were studied in 3.4.5. Zebrafish
European seabass (D. labrax) and Japanese seabass (L. japonicus) Zebrafish has all the enzymatic machinery enabling the biosynthesis
[110,111]. In Japanese seabass, a significant negative correlation was of LC-PUFA from the C18 precursors, LA and ALA, via the combined
observed between the CpG methylation of fads2 promoter and hepatic action of the gene products of Δ6Δ5 fads2 [23], elovl5 [69], elovl2 [94]
fads2 transcript levels in response to high dietary n-3 LC-PUFA (pre­ and elovl4 [86]. In recent years, the transcriptional regulation of LC-
dominantly DHA and EPA, ~2:1). However, the conserved NF-Y, SRE PUFA biosynthesis has been investigated in zebrafish with NF-Y,
and Sp1 sites were not methylated, and so the possible inhibition of CCAAT boxes, Sp1, SRE and PPRE binding sites recognized as impor­
fads2 expression by DNA methylation might be related to the inactive tant for maintaining and regulating Δ6Δ5 fads2 promoter activity [115],
chromatin structure and/or inhibition of another TF [111]. In European whereas NF-Y, SRE and Sp1 were vital for elovl5 promoter activity [119].
seabass, high dietary n-3 LC-PUFA (predominantly DHA and EPA, Moreover, Srebp-1 was shown to interact with SRE in the promoters of
~1.4:1) also reduced expression of Δ6 fads2, but no differences in Δ6Δ5 fads2 and elovl5, while NF-Y binds to CCAAT sites in the promoter
methylation at CpG sites were observed in the Δ6 fads2 promoter [110]. of Δ6Δ5 fads2 [115]. Overexpression of Srebp-1 in the zebrafish liver
Possible reasons for these different results might be the different DHA: cell line (ZFL ATCC® CRL-2643™) increased the activities of Δ6Δ5
EPA ratios, or n-3 LC-PUFA contents of the diets in the two studies with fads2 and elovl5 promoters, which provided further evidence of the
the high n-3 LC-PUFA diet (45 % of total fatty acids) being almost 30- transcriptional regulation mechanisms in D. rerio [115].
fold greater than that of the low n-3 LC-PUFA diet (1.5 % of total fatty
acids) in the Japanese seabass study, but the high n-3 LC-PUFA diet (1.2 3.4.6. Gilthead seabream
% of diet) was only 4-fold higher than the low diet (0.3 % of diet) in the Gilthead seabream is another marine teleost whose LC-PUFA
European seabass study [110,111]. biosynthesis pathway has attracted attention. The seabream fads2
gene was cloned and characterized with dual ∆6 and ∆8 desaturase ac­
3.4.3. Large yellow croaker tivity [25,35,148], and PUFA elongation capacity has been also
Evidence collected in large yellow croaker from in vivo feeding trials demonstrated through functional characterization of the Elovl5 and
and in vitro hepatocyte cell culture studies showed that levels of lxrα, Elovl4 elongases [70,90]. Like most marine fish, gilthead seabream has a

8
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

low capacity to biosynthesize LC-PUFA due to the apparent absence of investigated in the future.
Δ5 desaturase activity [35,148] and, thus, an adequate dietary supply of
LC-PUFA is critical for larval growth and development [149]. Although 4. The post-transcriptional regulation of LC-PUFA biosynthesis
several studies reported that Ppar, Sp1, Srebp-1, Lxrα, and Np-y may be in teleosts
involved in the regulation of energy metabolism in gilthead seabream
[150–152], no direct evidence supported the roles of these TF in LC- It is well known that most eukaryotic genes are also subject to
PUFA biosynthesis. However, feeding trial studies showed that the considerable post-transcriptional regulation, and thus transcribed
mRNA levels of pparγ, srebp-1, Δ6 fads2, and elovl4 increased in seab­ mRNA can undergo a variety of modifications including further pro­
ream fed low dietary LC-PUFA, which suggested that Pparγ and Srebp-1 cessing, export, localization, turnover, and translation, which are an
may play roles in regulating LC-PUFA biosynthesis [153,154]. Addi­ integral part of gene expression, and equally as sophisticated and
tionally, two recent studies showed that the promoters of stearoyl-CoA important as transcriptional control. Recent improvements in high
desaturase 1a and fads2 were responsive to broodstock nutrition, and throughput sequencing and computational prediction methods have
that both the parents fed VO diets (low n-3 LC-PUFA) and their offspring allowed the discovery of several types of non-coding RNA (ncRNA) that,
shared increased DNA-methylation [155,156], which revealed epige­ together with protein effector complexes, can also control the expression
netic mechanisms in the nutritional regulation of LC-PUFA biosynthesis of target mRNA at a post-transcriptional level [157]. Non-coding RNA,
in seabream. functional RNA molecules that are transcribed from DNA but do not
Overall, concerning transcriptional regulation of LC-PUFA biosyn­ code for proteins, includes microRNA (or miRNA), small interfering
thesis, similar regulatory mechanisms such as the Lxrα-Srebp-1 pathway RNA (siRNA), small nuclear RNA (snRNA) and long non-coding RNA
are conserved from teleosts to mammals. However, teleosts appear to (lncRNA) [157]. Among them, microRNA (or miRNA) appear as
possess more complex mechanisms of regulation as a result of the important post-transcriptional regulators of their mRNA targets via
increased gene repertoire and greater functional diversity of teleost mRNA degradation and/or translational repression [158]. MiRNA are
fads2 and, to some extent, elovl genes [20,34]. For example, Sp1 plays an highly conserved, small, ncRNA of 18–25 nt length that typically control
important role in determining the promoter activities of Δ6Δ5 fads2 and the expression of their targets by imperfect base pairing to the 3′ un­
elovl5 genes, Hnf4α and Pparα target widely the promoters of Δ6Δ5 translated regions (3′ UTR) of the target mRNA [159]. Now, miRNA have
fads2, Δ4 fads2, and elovl5 genes as a stimulatory (positive) TF, while emerged as crucial post-transcriptional regulators of lipid metabolism
Pparγ has inhibitory (negative) effects on Δ6Δ5 fads2 expression. A [160].
schematic overview of the transcriptional regulation of LC-PUFA While there have been no reports in other vertebrates, the roles of
biosynthesis in teleosts based on currently available data is shown in miRNA in the regulation of LC-PUFA biosynthesis has attracted attention
Fig. 3. However, this is far from a full understanding of transcriptional in teleosts. Recently, several miRNAs have been identified to be involved
regulation of LC-PUFA biosynthesis in teleosts. The influence of fish in LC-PUFA biosynthesis in the teleost model S. canaliculatus by target­
species, habitat and feeding habits, as well as impacts of salinity, tem­ ing LC-PUFA biosynthesis-related genes as outlined in Fig. 3. These
perature and dietary nutrients also require to be elucidated. Certainly, include miR-17 and miR-146a that directly target key desaturase and
expression of TF and downstream genes, and metabolism were changed elongase genes involved in LC-PUFA biosynthesis, and miR-24, miR-33,
with adaptation to environmental and dietary factors. Teleosts may miR-26a, miR-145 and the miR-15/16 cluster that target TF that, in turn,
respond to changes in these factors via stimuli acting through known cell regulate the expression of key desaturase and elongase genes involved in
signalling cascades, such as calcium ion [Ca2+] and kinase phosphory­ LC-PUFA biosynthesis [143,161–167]. Below, we provide a description
lation among others. However, presently little is known about these of the most important findings obtained in these studies.
cellular signalling pathways in the regulation of key enzyme genes
involved in LC-PUFA biosynthesis in fish, and this requires to be

Fig. 3. Schematic overview of transcription factors


(TF) and miRNA involved in the transcriptional and
post-transcriptional regulation of LC-PUFA biosyn­
thesis in teleosts. The main TF binding elements
identified in the core promoters of teleost fads2 and
elovl include GC rich region, nuclear factor Y (NF-Y),
sterol regulatory element (SRE), direct repeat 1 (DR-
1) and peroxisome proliferator response element
(PPRE). The TF include stimulatory protein 1 (Sp1),
sterol regulatory element-binding protein-1 (Srebp-
1), hepatocyte nuclear factor 4 alpha (Hnf4α), liver X
receptor alpha (Lxrα) and peroxisome proliferator-
activated receptors alpha/gamma (Pparα/γ). Several
miRNAs have been shown to be involved in LC-PUFA
biosynthesis in S. canaliculatus, where miR-17 and
miR-146a directly target Δ4 fads2 and elovl5,
respectively. miR-24 and miR-33 directly co-target
insulin-induced gene 1 (insig1), an endoplasmic re­
ticulum (ER) membrane protein that facilitates the
retention of Srebp-1 precursors in the ER and pre­
vents their proteolytic activation in the Golgi appa­
ratus. miR-26a, miR-145 and miR-15/16 cluster
target lxrα, hnf4α and pparγ, respectively. nSrebp-1,
the mature nuclear form of Srebp-1 protein; Scap,
Srebp cleavage-activating protein; TSS, transcrip­
tional start site. Pointed arrows: upregulation of
target mRNA expression. Blunt arrows: down-
regulation of target mRNA expression.

9
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

4.1. Inhibitory roles of miR-17, miR-146a, miR-26a and miR-145 in the its host, SREBP-1 [168]. Subsequently, miR-33 was investigated in
regulation of LC-PUFA biosynthesis rabbitfish and confirmed to be an intronic miRNA (intron 16) in the
srebp-1 gene [162]. Moreover, miR-33 was demonstrated to be involved
The first miRNA demonstrated to participate in the regulation of LC- in the regulation of LC-PUFA biosynthesis by cooperating with Srebp-1
PUFA biosynthesis in vertebrates was miR-17 in rabbitfish [161]. It is and/or through facilitating Srebp-1 processing by targeting insulin-
located in the forepart of the miR-17-92 cluster and its mature sequence induced gene 1 (Insig1) [143,162] (Fig. 3). INSIG1 is an endoplasmic
is highly consistent with that of other vertebrate species. In silico analysis reticulum (ER) membrane protein that facilitates the retention of
found a conserved complementary site for miR-17 in the 3′ UTR of rab­ SREBP-1 precursors in the ER and prevents their proteolytic activation
bitfish Δ4 fads2 mRNA, and dual luciferase reporter assays demon­ in the Golgi apparatus [169].
strated that miR-17 targeted the 3′ UTR of Δ4 fads2 directly [161]. Additionally, miR-24 was found to bind the 3′ UTR of insig1 mRNA in
Moreover, miR-17 abundance was significantly higher in the liver of rabbitfish [164]. Overexpression of miR-24 down-regulated Insig1
rabbitfish reared at 32 ppt salinity than at 10 ppt salinity, and showed protein level, and up-regulated mature Srebp-1 protein and gene
inverse trends with Δ4 fads2 gene expression and protein quantity. An expression of Δ4 fads2, Δ6Δ5 fads2 and elovl5 in SCHL cells. Knockdown
inverse expression pattern for miR-17 and Δ4 fads2 was also found in of miR-24 increased the expression of insig1 but inhibited the expression
rabbitfish primary hepatocytes incubated with 100 μM PUFA including of mature Srebp1 protein and Δ4 fads2, Δ6Δ5 fads2 and elovl5 genes,
ALA, EPA and DHA. These results demonstrated that miR-17 was and these effects could be attenuated by additional insig1 knockdown
involved in the regulation of LC-PUFA biosynthesis by targeting Δ4 (Fig. 3). Besides, knockdown of miR-24 in SCHL cells not only reduced
fads2 (Fig. 3). MUFA accumulation, but also suppressed LC-PUFA biosynthesis through
Similar to miR-17, another miRNA, miR-146a, was found to bind the inhibiting Insig1-dependent Srebp-1 activation and the expression of
3′ UTR of elovl5 that, as noted above, is one of the critical enzymes in the critical enzymes that are required for LC-PUFA biosynthesis [164].
elongation of C18 and C20 PUFA [5,13,65]. In vitro gain- and loss-of-
function experiments in SCHL cells demonstrated that miR-146a 4.3. Role of miRNA clusters in the regulation of LC-PUFA biosynthesis
decreased Elovl5-dependent PUFA elongation product/substrate
indices including 20:3n-6/18:3n-6, 20:4n-3/18:4n-3 and 22:5n-3/ Compared with individual miRNA, the regulatory model of miRNA
20:5n-3, as well as overall LC-PUFA contents, by inhibiting the expres­ clusters is more complex, and their function is more efficient [170,171].
sion of elovl5 [163] (Fig. 3). Recent studies demonstrated the role of the miR-15/16 cluster in the
Different to the regulatory mechanisms on LC-PUFA biosynthesis regulation of LC-PUFA biosynthesis in rabbitfish [167]. First, the
described above for miR-17 and miR-146a by directly targeting Δ4 fads2 abundance of miR-15/16 cluster was greatly increased in SCHL cells
and elovl5, respectively, miR-26a was found to exert its regulatory ef­ treated with the LC-PUFA biosynthetic precursor ALA, but was signifi­
fects on LC-PUFA biosynthesis indirectly by targeting the 3′ UTR of the cantly depressed by DHA and EPA, which suggested a possible role of the
TF lxrα, based on both in silico analysis and dual luciferase reporter as­ miR-15/16 cluster in LC-PUFA biosynthesis. Moreover, bioinformatic
says [165]. Overexpression of miR-26a in SCHL cells markedly reduced analysis showed a potential binding site shared for miR-15 and miR-16
the protein levels of Lxrα, Srebp-1 and Δ6Δ5 Fads2 induced by the Lxr in the 3′ UTR of pparγ, a negative regulator of LC-PUFA biosynthesis in
agonist T0901317. On the contrary, knockdown of miR-26a using rabbitfish as mentioned above. In vitro, luciferase reporter assays
antagomirs (i.e. anti-MiR) facilitated Srebp-1 processing and increased revealed that pparγ was a potential target of the miR-15/16 cluster, and
the expression of Δ6Δ5fads2, Δ4 fads2 and elovl5 genes involved in LC- individual or co-overexpression of miR-15 and miR-16 in SCHL cells
PUFA biosynthesis, and consequently, promoted LC-PUFA biosynthesis significantly inhibited both mRNA and protein levels of Pparγ, but
in both hepatocytes in vitro and rabbitfish in vivo. Combining the results increased the mRNA levels of Δ6Δ5 fads2, Δ4 fads2 and elovl5. This led
on miR-26a and those of the role of the Lxrα-Srebp-1 pathway in the to increased contents of DHA and ARA in the SCHL cells with higher
regulation of LC-PUFA biosynthesis as mentioned above (Section 3.3) product/substrate ratios for Δ6Δ5 and Δ4 desaturation activities.
[162], it has been suggested that miR-26a plays a critical role in regu­ Furthermore, inhibition of Pparγ was more pronounced with co-
lating LC-PUFA biosynthesis through targeting the Lxrα-Srebp-1 overexpression of miR-15 and miR-16 than with individual over­
pathway in rabbitfish [165] (Fig. 3). expression in SCHL cells. Consistently, knockdown of the miR-15/16
Similarly, miR-145 was found to target the 3′ UTR of hnf4α that, as cluster gave the opposite results, and increased mRNA levels of LC-
noted above, is an important TF involved in LC-PUFA biosynthesis in PUFA biosynthesis enzymes were observed after knockdown of pparγ.
rabbitfish by regulating expression of ∆4 fads2, Δ6Δ5 fads2 and elovl5 These gain- and loss-of-function experiments demonstrated that miR-15
[114,116,120,126,142]. In vitro gain- and loss-of-function experiments and miR-16 act as a miRNA cluster to enhance LC-PUFA biosynthesis by
in SCHL cells demonstrated that miR-145 decreased the expression of ∆4 targeting Pparγ in rabbitfish, uncovering a novel mechanism that may
fads2, Δ6Δ5 fads2 and elovl5 genes, and consequently reduced total LC- also operate in other vertebrates [167].
PUFA contents by targeting hnf4α [166]. Moreover, knockdown of miR- Overall, studies on the roles of miRNA in the regulation of LC-PUFA
145 by intraperitoneal injection with antagomirs increased Hnf4α pro­ biosynthesis have only been reported in S. canaliculatus, but not yet in
tein level and its downstream target genes expression (Δ6Δ5fads2, Δ4 other fish species or other vertebrates, except for miR-193a-5p that was
fads2 and elovl5), as well as total LC-PUFA contents in fish tissues, shown to regulate LC-PUFA biosynthesis by targeting FADS1 in bovine
including liver, brain and eyes, where LC-PUFA biosynthetic activity is mammary epithelial cells [172]. As described above, individual miRNA
particularly high in fish [83]. These results revealed that miR-145 may or clusters regulated rabbitfish LC-PUFA biosynthesis by targeting fads2,
be a key mediator involved in the regulation of LC-PUFA biosynthesis elovl or TF-encoding genes. At present, little is known about how
both in vivo and in vitro through targeting hnf4α in the marine teleost external factors such as ambient salinity, temperature, or dietary nu­
S. canaliculatus [166] (Fig. 3). trients like fatty acids are signaled to cellular cascades to affect miRNA
that can then influence TF with the latter subsequently regulating
4.2. Stimulatory roles of miR-33 and miR-24 in the regulation of LC- enzyme gene transcription or protein translation and, eventually,
PUFA biosynthesis enzymic activities. Moreover, it is not known whether feedback and
feed-forward loops are present in teleosts. In these, a TF regulates a
In mammals, miR-33 has been found within an intron of SREBP-1, an miRNA or a miRNA modulates a TF, with both co-regulating the same
important TF involved in regulating the expression of genes encoding target genes involved in LC-PUFA biosynthesis as reported in mammals
key enzymes of vertebrate LC-PUFA biosynthesis and, consequently, where these regulatory loops are important regulatory motifs in multiple
miR-33 was reported to modulate lipid metabolism in cooperation with biological processes [173]. Thus, it is crucial to identify and study

10
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

miRNA-TF-gene or TF-miRNA-gene co-regulatory networks in teleosts as related to LC-PUFA biosynthesis in different fish species also increases
the application of various genome-wide approaches increases in the the difficulties in investigating the multifaceted control of gene
future so as to provide a more comprehensive insight into the regulatory expression at the post-transcriptional level. Studies of post-
mechanisms of LC-PUFA biosynthesis. transcriptional control by miRNA have provided a glimpse into the
richness and sophistication of mRNA regulation. As the application of
5. Conclusions and perspective various genome-wide approaches increases, the next decade will bring
an even more comprehensive view of all levels of the cellular regulation
In recent years, the regulatory mechanisms of LC-PUFA biosynthesis of LC-PUFA biosynthesis in teleosts.
in teleost fish has become a hot research topic in lipid nutrition, as it
provides insight into the metabolic responses that are triggered in fish Author contributions
when fed diets with high inclusion of VO that dominate the market in
intensive aquaculture. The fundamental biochemical and molecular Dizhi Xie, Cuiying Chen, Yewei Dong and Shuqi Wang: Conceptual­
studies will facilitate the development of practical strategies that can ization, writing original draft; Cuihong You, Óscar Monroig and Douglas
enable increased replacement of dietary FO (rich in LC-PUFA) with VO R. Tocher: Writing, review and editing of manuscript; Yuanyou Li:
(rich in C18 PUFA precursors, but devoid of LC-PUFA) in farmed teleost Writing, design, review and editing, supervision and funding acquisi­
diets, while ensuring that this does not negatively impact the health of tion. All authors approved the submission and publication of this
the fish or its nutritional quality (i.e. content of n-3 LC-PUFA) as human manuscript.
food [174].
There are at least five TF involved in the transcriptional regulation of
LC-PUFA biosynthesis in teleosts, with Hnf4α, Lxrα-Srebp-1 and Sp1 Declaration of Competing Interest
displaying positive/stimulatory roles, and Pparγ playing negative/
inhibitory effects. Indeed, evidence obtained in S. canaliculatus was the None.
first demonstration that Hnf4α and Pparγ have major regulatory func­
tions in LC-PUFA biosynthesis and provided clarification of the impor­ Acknowledgements
tant role of Sp1 in determining the transcriptional control of genes
involved LC-PUFA biosynthesis in vertebrates. With respect to post- This work was financially supported by the National Key R&D Pro­
transcriptional regulation of LC-PUFA biosynthesis, studies showed gram of China (2018YFD0900400) and National Natural Science
that specific miRNA or miRNA clusters play important roles by acting Foundation of China (31873040, 31110103913, 30972266, 30671629,
either directly on key fads and elovl genes involved in LC-PUFA 31702357), and partly funded through the project IMPROMEGA of the
biosynthesis or indirectly by regulating TF that, in turn, themselves Ministry of Science, Innovation and Universities, Spanish Government
modulate the expression of the fads and elovl genes. While miR-33, miR- (grant no. RTI2018-095119-B-I00, MCIU/AEI/FEDER, UE).
24 and the miR-15/16 cluster have stimulatory roles in LC-PUFA
biosynthesis in teleosts, miR-17, miR-26a, miR-145 and miR-146a References
have inhibitory roles. Fig. 3 summarizes the regulatory mechanisms of
LC-PUFA biosynthesis in teleosts, showing several aspects with complex [1] Vannice G, Rasmussen H. Position of the academy of nutrition and dietetics:
dietary fatty acids for healthy adults. J Acad Nutr Diet 2014;114:136–53. https://
interactions. Among them, Srebp-1 and Hnf4α might play more central doi.org/10.1016/j.jand.2013.11.001.
roles in the regulation of LC-PUFA biosynthesis. For example, Lxrα, miR- [2] Keim SA, Branum AM. Dietary intake of polyunsaturated fatty acids and fish
33, miR-24 and Insig1 regulated LC-PUFA biosynthesis via Srebp-1, among US children 12-60 months of age. Matern Child Nutr 2015;11:987–98.
https://doi.org/10.1111/mcn.12077.
while the regulation of Sp1 on key enzymes of LC-PUFA biosynthesis [3] FAO. The state of world fisheries and aquaculture 2020: Sustainability in action.
also depends on Srebp-1. Other factors, such as Pparγ, miR-17, miR-26a, Rome, Italy: Food and Agriculture Organisation of the United Nations; 2020.
miR-145, miR-146a and miR-15/16 directly or indirectly target different p. 9–65. https://doi.org/10.4060/ca9229en.
[4] Subasinghe R, Soto D, Jia J. Global aquaculture and its role in sustainable
nodes to form a complex network to regulate LC-PUFA biosynthesis. development. Rev Aquacult 2009;1:2–9. https://doi.org/10.1111/j.1753-
While rabbitfish is a robust model for the investigation of LC-PUFA 5131.2008.01002.x.
biosynthesis in vertebrates, a major challenge that needs to be [5] Monroig Ó, Tocher DR, Castro LFC. Polyunsaturated fatty acid biosynthesis and
metabolism in fish. In: Burdge GC, editor. Polyunsaturated fatty acid metabolism.
addressed in the future is the sequencing of the rabbitfish genome as­
London: Academic Press and AOCS Press; 2018. p. 31–60. https://doi.org/
sembly that will enable, among others, the application of gene knockout 10.1016/B978-0-12-811230-4.00003-X.
methodologies (e.g. CRISPR/Cas9), nowadays regarded as the “gold [6] Naylor RL, Hardy RW, Bureau DP, Chiu A, Elliott M, Farrell AP, et al. Feeding
standard” in gene function analyses. Further studies are also required to aquaculture in an era of finite resources. Proc Natl Acad Sci USA 2009;106:
15103–10. https://doi.org/10.1073/pnas.0905235106.
understand how environmental factors such as salinity, and nutritional [7] Tocher DR, Betancor MB, Sprague M, Olsen RE, Napier JA. Omega-3 long-chain
factors such as lipids and fatty acids, affect LC-PUFA biosynthesis. polyunsaturated fatty acids, EPA and DHA: bridging the gap between supply and
Current data collected through the studies presented above suggest that demand. Nutrients 2019;11:89. https://doi.org/10.3390/nu11010089.
[8] Tocher DR, Sargent JR. Effect of temperature on the incorporation into
miRNA play pivotal roles in the metabolic responses of teleosts to phospholipid classes and metabolism via desaturation and elongation of n-3 and
environmental challenges affecting lipid homeostasis. Besides, the reg­ n-6 polyunsaturated fatty acids in fish cells in culture. Lipids 1990;25:435–42.
ulatory network of LC-PUFA biosynthesis in teleosts needs to be further https://doi.org/10.1007/BF02538085.
[9] Rosenlund G, Torstensen BE, Stubhaug I, Usman N, Sissener NH. Atlantic salmon
investigated and integrated within the overall regulation of various lipid require long-chain n-3 fatty acids for optimal growth throughout the seawater
metabolic pathways. The regulation of both de novo lipogenesis and LC- period. J Nutr Sci 2016;5:e19. https://doi.org/10.1017/jns.2016.10.
PUFA biosynthesis by Srebp-1 and Hnf4α, suggests a close relationship [10] Xu H, Turchini GM, Francis DS, Liang M, Mock TS, Rombenso A, et al. Are fish
what they eat? A fatty acid’s perspective. Prog Lipid Res 2020:101064. https://
between the different metabolic pathways, which indicates that we must doi.org/10.1016/j.plipres.2020.101064.
consider more aspects when exploring new mechanisms of lipid meta­ [11] Castro LFC, Monroig Ó, Leaver MJ, Wilson J, Cunha I, Tocher DR. Functional
bolism, including different pathways and different tissues. This can be desaturase Fads1 (Δ5) and Fads2 (Δ6) orthologues evolved before the origin of
jawed vertebrates. PLoS One 2012;7:e31950. https://doi.org/10.1371/journal.
achieved by systematic analysis of the transcriptome, proteome, and
pone.0031950.
metabolome. [12] Oboh A, Kabeya N, Carmona-Antoñanzas G, Castro LFC, Dick JR, Tocher DR,
Until now, there has been relatively little research into the post- et al. Two alternative pathways for docosahexaenoic acid (DHA, 22:6n-3)
transcriptional regulation of LC-PUFA biosynthesis in other fish spe­ biosynthesis are widespread among teleost fish. Sci Rep 2017;7:1–10. https://doi.
org/10.1038/s41598-017-04288-2.
cies. The substantial complexity of gene expression after transcription [13] Castro LFC, Tocher DR, Monroig Ó. Long-chain polyunsaturated fatty acid
may be one of the reasons. Lack of commercial antibodies for proteins biosynthesis in chordates: Insights into the evolution of Fads and Elovl gene

11
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

repertoire. Prog Lipid Res 2016;62:25–40. https://doi.org/10.1016/j. [35] Zheng X, Seiliez I, Hastings N, Tocher DR, Panserat S, Dickson CA, et al.
plipres.2016.01.001. Characterization and comparison of fatty acyl Δ6 desaturase cDNAs from
[14] Pereira SL, Leonard AE, Mukerji P. Recent advances in the study of fatty acid freshwater and marine teleost fish species. Comp Biochem Phys B 2004;139:
desaturases from animals and lower eukaryotes. Prostag Leukotr Ess 2003;68: 269–79. https://doi.org/10.1016/j.cbpc.2004.08.003.
97–106. https://doi.org/10.1016/S0952-3278(02)00259-4. [36] Xie D, Ye J, Lu M, Wang S, You C, Li Y. Comparison of activities of fatty acyl
[15] Ruiz-Lopez N, Usher S, Sayanova OV, Napier JA, Haslam RP. Modifying the lipid desaturases and elongases among six teleosts with different feeding habits and
content and composition of plant seeds: engineering the production of LC-PUFA. ecological habits. Front Mar Sci 2020;7:117. https://doi.org/10.3389/
Appl Microbiol Biot 2015;99:143–54. https://doi.org/10.1007/s00253-014- fmars.2020.00117.
6217-2. [37] Geay F, Tinti E, Mellery J, Michaux C, Larondelle Y, Perpète E, et al. Cloning and
[16] Kabeya N, Fonseca MM, Ferrier DEK, Navarro JC, Bay LK, Francis DS, et al. Genes functional characterization of Δ6 fatty acid desaturase (FADS2) in Eurasian perch
for de novo biosynthesis of omega-3 polyunsaturated fatty acids are widespread in (Perca fluviatilis). Comp Biochem Phys B 2016;191:112–25. https://doi.org/
animals. Sci Adv 2018;4:eaar6849. https://doi.org/10.1126/sciadv.aar6849. 10.1016/j.cbpb.2015.10.004.
[17] Wang S, Wang M, Zhang H, Yan X, Guo H, You C, et al. Long-chain [38] Lopes-Marques M, Ozório R, Amaral R, Tocher DR, Monroig Ó, et al. Molecular
polyunsaturated fatty acid metabolism in carnivorous marine teleosts: insight into and functional characterization of a fads2 orthologue in the Amazonian teleost,
the profile of endogenous biosynthesis in golden pompano Trachinotus ovatus. Arapaima gigas. Comp Biochem Phys B 2017;203:84–91. https://doi.org/
Aquac Res 2020;51:623–35. https://doi.org/10.1111/are.14410. 10.1016/j.cbpb.2016.09.007.
[18] Sprecher H. Metabolism of highly unsaturated n-3 and n-6 fatty acids. BBA-Mol [39] Monroig Ó, Tocher DR, Hontoria F, Navarro JC. Functional characterisation of a
Cell Biol L 2000;1486:219–31. https://doi.org/10.1016/S1388-1981(00)00077- Fads2 fatty acyl desaturase with Δ6/Δ8 activity and an Elovl5 with C16, C18 and
9. C20 elongase activity in the anadromous teleost meagre (Argyrosomus regius).
[19] Li Y, Monroig Ó, Zhang L, Wang S, Zheng X, Dick JR, et al. Vertebrate fatty acyl Aquaculture 2013;412:14–22. https://doi.org/10.1016/j.
desaturase with Δ4 activity. Proc Natl Acad Sci USA 2010;107:16840–5. https:// aquaculture.2013.06.032.
doi.org/10.1073/pnas.1008429107. [40] Kim SH, Roh KH, Kim JB, Kim KS, Kim NS, Kim HU, et al. Isolation and functional
[20] Lopes-Marques M, Kabeya N, Qian Y, Ruivo R, Santos MM, Venkatesh B, et al. characterization of a delta 6-desaturase gene from the pike eel (Muraenesox
Retention of fatty acyl desaturase 1 (fads1) in Elopomorpha and Cyclostomata cinereus). J Microbiol 2013;51:807–13. https://doi.org/10.1007/s12275-013-
provides novel insights into the evolution of long-chain polyunsaturated fatty 3144-3.
acid biosynthesis in vertebrates. BMC Evol Biol 2018;18:157. https://doi.org/ [41] Wang S, Monroig Ó, Tang G, Zhang L, You C, Tocher DR, et al. Investigating long-
10.1186/s12862-018-1271-5. chain polyunsaturated fatty acid biosynthesis in teleost fish: functional
[21] Hastings N, Agaba MK, Tocher DR, Zheng X, Dickson CA, Dick JR, et al. Molecular characterization of fatty acyl desaturase (Fads2) and Elovl5 elongase in the
cloning and functional characterization of fatty acyl desaturase and elongase catadromous species, Japanese eel Anguilla japonica. Aquaculture 2014;434:
cDNAs involved in the production of eicosapentaenoic and docosahexaenoic acids 57–65. https://doi.org/10.1016/j.aquaculture.2014.07.016.
from α-linolenic acid in Atlantic salmon (Salmo salar). Mar Biotechnol 2004;6: [42] Morais S, Mourente G, Ortega A, Tocher JA, Tocher DR. Expression of fatty acyl
463–74. https://doi.org/10.1007/s10126-004-3002-8. desaturase and elongase genes, and evolution of DHA: EPA ratio during
[22] Monroig Ó, Zheng X, Morais S, Leaver MJ, Taggart JB, Tocher DR. Multiple genes development of unfed larvae of Atlantic bluefin tuna (Thunnus thynnus L.).
for functional ∆6 fatty acyl desaturases (Fad) in Atlantic salmon (Salmo salar L.): Aquaculture 2011;313:129–39. https://doi.org/10.1016/j.
gene and cDNA characterization, functional expression, tissue distribution and aquaculture.2011.01.031.
nutritional regulation. BBA-Mol Cell Biol L 2010;1801:1072–81. https://doi.org/ [43] Tocher DR, Zheng X, Schlechtriem C, Hastings N, Dick JR, Teale AJ. Highly
10.1016/j.bbalip.2010.04.007. unsaturated fatty acid synthesis in marine fish: cloning, functional
[23] Hastings N, Agaba M, Tocher DR, Leaver MJ, Dick JR, Sargent JR, et al. characterization, and nutritional regulation of fatty acyl delta 6 desaturase of
A vertebrate fatty acid desaturase with Delta 5 and Delta 6 activities. Proc Natl Atlantic cod (Gadus morhua L.). Lipids 2006;41:1003–16. https://doi.org/
Acad Sci USA 2001;98:14304–9. https://doi.org/10.1073/pnas.251516598. 10.1007/s11745-006-5051-4.
[24] Park WJ, Kothapalli KS, Lawrence P, Tyburczy C, Brenna JT. An alternate [44] Tu WC, Cook-Johnson RJ, James MJ, Mühlhäusler BS, Stone DA, Gibson RA.
pathway to long-chain polyunsaturates: the FADS2 gene product delta8- Barramundi (Lates calcarifer) desaturase with Δ6/Δ8 dual activities. Biotechnol
desaturates 20:2n-6 and 20:3n-3. J Lipid Res 2009;50:1195–202. https://doi.org/ Lett 2012;34:1283–96. https://doi.org/10.1007/s10529-012-0891-x.
10.1194/jlr.M800630-JLR200. [45] Kim SH, Park JS, Kim SY, Kim JB, Roh KH, Kim HU, et al. Functional
[25] Monroig Ó, Li Y, Tocher DR. Delta-8 desaturation activity varies among fatty acyl characterization of polyunsaturated fatty acid delta 6-desaturase and elongase
desaturases of teleost fish: high activity in delta-6 desaturases of marine species. genes from the black seabream (Acanthopagrus schlegelii). Cell Biochem Biophys
Comp Biochem Phys B 2011;159:206–13. https://doi.org/10.1016/j. 2014;68:335–46. https://doi.org/10.1007/s12013-013-9714-5.
cbpb.2011.04.007. [46] Huang Y, Lin Z, Rong H, Hao M, Zou W, Li S, et al. Cloning, tissue distribution,
[26] Sprecher H, Luthria DL, Mohammed BS, Baykousheva SP. Reevaluation of the functional characterization and nutritional regulation of Δ6 fatty acyl desaturase
pathways for the biosynthesis of polyunsaturated fatty acids. J Lipid Res 1995;36: in Chu’s croaker Nibea coibor. Aquaculture 2017;479:208–16. https://doi.org/
2471–7. http://www.jlr.org/cgi/content/abstract/36/12/2471. 10.1016/j.aquaculture.2017.05.041.
[27] Buzzi M, Henderson RJ, Sargent JR. Biosynthesis of docosahexaenoic acid in trout [47] Zheng X, Ding Z, Xu Y, Monroig Ó, Morais S, Tocher DR. Physiological roles of
hepatocytes proceeds via 24-carbon intermediates. Comp Biochem Phys B 1997; fatty acyl desaturases and elongases in marine fish: characterisation of cDNAs of
116:263–7. https://doi.org/10.1016/S0305-0491(96)00210-6. fatty acyl Δ6 desaturase and elovl5 elongase of cobia (Rachycentron canadum).
[28] Tocher DR, Agaba M, Hastings N, Teale AJ. Biochemical and molecular studies of Aquaculture 2009;290:122–31. https://doi.org/10.1016/j.
the fatty acid desaturation pathway in fish. In: Browman HI, Skiftesvik AB, aquaculture.2009.02.010.
editors. The big fish bang: proceedings of the 26th annual larval fish conference. [48] Li S, Mai K, Xu W, Yuan Y, Zhang Y, Ai Q. Characterization, mRNA expression and
Bergen, Norway: Institute of Marine Nutrition; 2003. p. 211–27. http://hdl. regulation of Δ6 fatty acyl desaturase (FADS2) by dietary n-3 long chain
handle.net/1893/2896. polyunsaturated fatty acid (LC-PUFA) levels in grouper larvae (Epinephelus
[29] Betancor MB, Oboh A, Ortega A, Mourente G, Navarro JC, de la Gandara F, et al. coioides). Aquaculture 2014;434:212–9. https://doi.org/10.1016/j.
Molecular and functional characterisation of a putative elovl4 gene and its aquaculture.2014.08.009.
expression in response to dietary fatty acid profile in Atlantic bluefin tuna [49] Kabeya N, Chiba M, Haga Y, Satoh S, Yoshizaki G. Cloning and functional
(Thunnus thynnus). Comp Biochem Phys B 2020;240:110372. https://doi.org/ characterization of fads2 desaturase and elovl5 elongase from Japanese flounder
10.1016/j.cbpb.2019.110372. Paralichthys olivaceus. Comp Biochem Phys B 2017;214:36–46. https://doi.org/
[30] Morais S, Castanheira F, Martinez-Rubio L, Conceição LE, Tocher DR. Long chain 10.1016/j.cbpb.2017.09.002.
polyunsaturated fatty acid synthesis in a marine vertebrate: ontogenetic and [50] Kabeya N, Yamamoto Y, Cummins SF, Elizur A, Yazawa R, Takeuchi Y, et al.
nutritional regulation of a fatty acyl desaturase with Δ4 activity. BBA-Mol Cell Polyunsaturated fatty acid metabolism in a marine teleost, Nibe croaker Nibea
Biol L 2012;1821:660–71. https://doi.org/10.1016/j.bbalip.2011.12.011. mitsukurii: Functional characterization of Fads2 desaturase and Elovl5 and Elovl4
[31] Fonseca-Madrigal J, Navarro JC, Hontoria F, Tocher DR, Martínez-Palacios CA, elongases. Comp Biochem Phys B 2015;188:37–45. https://doi.org/10.1016/j.
Monroig Ó. Diversification of substrate specificities in teleostei Fads2: cbpb.2015.06.005.
characterization of Δ4 and Δ6Δ5 desaturases of Chirostoma estor. J Lipid Res [51] Garrido D, Kabeya N, Betancor MB, Pérez JA, Acosta NG, Tocher DR, et al.
2014;55:1408–19. https://doi.org/10.1194/jlr.M049791. Functional diversification of teleost Fads2 fatty acyl desaturases occurs
[32] Kuah MK, Jaya-Ram A, Shu-Chien AC. The capacity for long-chain independently of the trophic level. Sci Rep 2019;9:1–10. https://doi.org/
polyunsaturated fatty acid synthesis in a carnivorous vertebrate: functional 10.1038/s41598-019-47709-0.
characterisation and nutritional regulation of a Fads2 fatty acyl desaturase with [52] Zou W, Lin Z, Huang Y, Limbu S, Wen X. Molecular cloning and functional
Δ4 activity and an Elovl5 elongase in striped snakehead (Channa striata). BBA characterization of elongase (elovl5) and fatty acyl desaturase (fads2) in sciaenid,
Mol Cell Biol L 2015;1851:248–60. https://doi.org/10.1016/j. Nibea diacanthus (Lacepède, 1802). Gene 2019;695:1–11. https://doi.org/
bbalip.2014.12.012. 10.1016/j.gene.2019.01.033.
[33] Galindo A, Garrido D, Monroig Ó, Perez JA, Betancor MB, Acosta NG, et al. [53] Santigosa E, Geay F, Tonon T, Le Delliou H, Kuhl H, Reinhardt R, et al. Cloning,
Polyunsaturated fatty acid metabolism in three fish species with different trophic tissue expression analysis, and functional characterization of two Δ6-desaturase
level. Aquaculture 2020;530:735761. https://doi.org/10.1016/j. variants of sea bass (Dicentrarchus labrax L.). Mar Biotechnol 2011;13:22–31.
aquaculture.2020.735761. https://doi.org/10.1007/s10126-010-9264-4.
[34] Guillou H, Zadravec D, Martin PG, Jacobsson A. The key roles of elongases and [54] Xie D, Chen F, Lin S, Wang S, You C, Monroig Ó, et al. Cloning, functional
desaturases in mammalian fatty acid metabolism: insights from transgenic mice. characterization and nutritional regulation of Δ6 fatty acyl desaturase in the
Prog Lipid Res 2010;49:186–99. https://doi.org/10.1016/j.plipres.2009.12.002. herbivorous euryhaline teleost Scatophagus argus. PLoS One 2014;9:e90200.
https://doi.org/10.1371/journal.pone.0090200.

12
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

[55] Soo HJ, Chong J, Lau NS, Ting SY, Sam KK, Kuah MK, et al. Functional [77] Mohd-Yusof NY, Monroig Ó, Mohd-Adnan A, Wan KL, Tocher DR. Investigation of
characterization of fatty acyl desaturase Fads2 and Elovl5 elongase in the highly unsaturated fatty acid metabolism in the Asian sea bass, Lates calcarifer.
Boddart’s goggle-eyed goby, Boleophthalmus boddarti (Gobiidae) suggest an Fish Physiol Biochem 2010;36:827–43. https://doi.org/10.1007/s10695-010-
incapacity for long-chain polyunsaturated fatty acid biosynthesis. J Fish Biol 9409-4.
2020;97:83–99. https://doi.org/10.1111/jfb.14328. [78] Kim SH, Kim JB, Jang YS, Kim SY, Roh KH, Kim HU, et al. Isolation and functional
[56] Abdul Hamid NK, Carmona-Antoñanzas G, Monroig Ó, Tocher DR, Turchini GM, characterization of polyunsaturated fatty acid elongase (AsELOVL5) gene from
Donald JA. Isolation and functional characterisation of a fads2 in rainbow trout black seabream (Acanthopagrus schlegelii). Biotechnolo Lett 2012;34:261–8.
(Oncorhynchus mykiss) with Δ5 desaturase activity. PLoS One 2016;11:e0150770. https://doi.org/10.1007/s10529-011-0746-x.
https://doi.org/10.1371/journal.pone.0150770. [79] Lin Z, Huang Y, Zou W, Rong H, Hao M, Wen X. Cloning, tissue distribution,
[57] Oboh A, Betancor MB, Tocher DR, Monroig Ó. Biosynthesis of long-chain functional characterization and nutritional regulation of a fatty acyl Elovl5
polyunsaturated fatty acids in the African catfish Clarias gariepinus: Molecular elongase in Chu’s croaker Nibea coibor. Gene 2018;659:11–21. https://doi.org/
cloning and functional characterisation of fatty acyl desaturase (fads2) and 10.1016/j.gene.2018.03.046.
elongase (elovl2) cDNAs. Aquaculture 2016;462:70–9. https://doi.org/10.1016/j. [80] Zhu K, Song L, Zhao C, Guo H, Zhang N, Guo L, et al. The transcriptional factor
aquaculture.2016.05.018. PPARαb positively regulates Elovl5 elongase in golden pompano Trachinotus
[58] Janaranjani M, Mah MQ, Kuah MK, Fadhilah N, Hing SR, Han WY, et al. Capacity ovatus (Linnaeus 1758). Front Physiol 2018;9:1340. https://doi.org/10.3389/
for eicosapentaenoic acid and arachidonic acid biosynthesis in silver barb fphys.2018.01340.
(Barbonymus gonionotus): functional characterisation of a Δ6/Δ8/Δ5 Fads2 [81] Li S, Monroig Ó, Wang T, Yuan Y, Navarro JC, Hontoria F, et al. Functional
desaturase and Elovl5 elongase. Aquaculture 2018;497:469–86. https://doi.org/ characterization and differential nutritional regulation of putative Elovl5 and
10.1016/j.aquaculture.2018.08.019. Elovl4 elongases in large yellow croaker (Larimichthys crocea). Sci Rep UK 2017;7:
[59] Ferraz RB, Kabeya N, Lopes-Marques M, Machado AM, Ribeiro RA, Salaro AL, 1–15. https://doi.org/10.1038/s41598-017-02646-8.
et al. A complete enzymatic capacity for long-chain polyunsaturated fatty acid [82] Li S, Yuan Y, Wang T, Xu W, Li M, Mai K, et al. Molecular cloning, functional
biosynthesis is present in the Amazonian teleost tambaqui, Colossoma characterization and nutritional regulation of the putative elongase Elovl5 in the
macropomum. Comp Biochem Phys B 2019;227:90–7. https://doi.org/10.1016/j. orange-spotted grouper (Epinephelus coioides). PloS One 2016;11:e0150544.
cbpb.2018.09.003. https://doi.org/10.1371/journal.pone.0150544.
[60] Garrido D, Monroig Ó, Galindo A, Betancor MB, Pérez JA, Kabeya N, et al. Lipid [83] Monroig Ó, Wang S, Zhang L, You C, Tocher DR, Li Y. Elongation of long-chain
metabolism in Tinca tinca and its n-3 LC-PUFA biosynthesis capacity. Aquaculture fatty acids in rabbitfish Siganus canaliculatus: Cloning, functional characterisation
2020;523:735147. https://doi.org/10.1016/j.aquaculture.2020.735147. and tissue distribution of Elovl5-and Elovl4-like elongases. Aquaculture 2012;
[61] Kuah MK, Jaya-Ram A, Shu-Chien AC. A fatty acyl desaturase (fads2) with dual 350:63–70. https://doi.org/10.1016/j.aquaculture.2012.04.017.
Δ6 and Δ5 activities from the freshwater carnivorous striped snakehead Channa [84] Gregory MK, See VH, Gibson RA, Schuller KA. Cloning and functional
striata. Comp Biochem Physiol A 2016;201:146–55. https://doi.org/10.1016/j. characterisation of a fatty acyl elongase from southern bluefin tuna (Thunnus
cbpa.2016.07.007. maccoyii). Comp Biochem Physiol B 2010;155:178–85. https://doi.org/10.1016/
[62] Zhu K, Zhang N, Liu B, Guo L, Guo H, Jiang S, et al. Transcription factor pparαb j.cbpb.2009.11.002.
activates fads2s to promote LC-PUFA biosynthesis in the golden pompano [85] Xie D, Chen F, Lin S, You C, Wang S, Zhang Q, et al. Long-chain polyunsaturated
Trachinotus ovatus (Linnaeus 1758). Int J Biol Macromol 2020;15:605–16. fatty acid biosynthesis in the euryhaline herbivorous teleost Scatophagus argus:
https://doi.org/10.1016/j.ijbiomac.2020.06.085. functional characterization, tissue expression and nutritional regulation of two
[63] Ishikawa A, Kabeya N, Ikeya K, Kakioka R, Cech JN, Osada N, et al. A key fatty acyl elongases. Comp Biochem Physiol B 2016;198:37–45. https://doi.org/
metabolic gene for recurrent freshwater colonization and radiation in fishes. 10.1016/j.cbpb.2016.03.009.
Science 2019;364:886–9. https://doi.org/10.1126/science.aau5656. [86] Monroig Ó, Rotllant J, Cerdá-Reverter JM, Dick JR, Figueras A, Tocher DR.
[64] Matsushita Y, Miyoshi K, Kabeya N, Sanaka S, Yazawa R, Haga Y, et al. Flatfishes Expression and role of Elovl4 elongases in biosynthesis of very long-chain fatty
colonised freshwater environments by acquisition of various DHA biosynthetic acids during zebrafish Danio rerio early embryonic development. BBA Mol Cell
pathways. Comms Biol 2020;3:516. https://doi.org/10.1038/s42003-020-01242- Biol L 2010;1801:1145–54. https://doi.org/10.1016/j.bbalip.2010.06.005.
3. [87] Yan J, Liang X, Cui Y, Cao X, Gao J. Elovl4 can effectively elongate C18
[65] Jakobsson A, Westerberg R, Jacobsson A. Fatty acid elongases in mammals: their polyunsaturated fatty acids in loach Misgurnus anguillicaudatus. Biochem Bioph
regulation and roles in metabolism. Prog Lipid Res 2006;45:237–49. https://doi. Res Co 2018;495:2637–42. https://doi.org/10.1016/j.bbrc.2017.12.123.
org/10.1016/j.plipres.2006.01.004. [88] Carmona-Antoñanzas G, Monroig Ó, Dick JR, Davie A, Tocher DR. Biosynthesis of
[66] Yamashita Y, Nishiumi S, Kono S, Takao S, Azuma T, Yoshida M. Differences in very long-chain fatty acids (C > 24) in Atlantic salmon: cloning, functional
elongation of very long chain fatty acids and fatty acid metabolism between characterisation, and tissue distribution of an Elovl4 elongase. Comp Biochem
triple-negative and hormone receptor-positive breast cancer. BMC Cancer 2017; Physiol B 2011;159:122–9. https://doi.org/10.1016/j.cbpb.2011.02.007.
17:589. https://doi.org/10.1186/s12885-017-3554-4. [89] Monroig Ó, Webb K, Ibarra-Castro L, Holt GJ, Tocher DR. Biosynthesis of long-
[67] Burdge GC. Biochemistry and regulation of Elongases 2 and 5 in mammals. In: chain polyunsaturated fatty acids in marine fish: characterization of an Elovl4-
Polyunsaturated fatty acid metabolism. AOCS Press; 2018. p. 101–9. https://doi. like elongase from cobia Rachycentron canadum and activation of the pathway
org/10.1016/B978-0-12-811230-4.00006-5. during early life stages. Aquaculture 2011;312:145–53. https://doi.org/10.1016/
[68] Hopiavuori BR, Anderson RE, Agbaga MP. ELOVL4: very long-chain fatty acids j.aquaculture.2010.12.024.
serve an eclectic role in mammalian health and function. Prog Retin Eye Res [90] Morais S, Torres M, Hontoria F, Monroig Ó, Varó I, Agulleiro MJ, et al. Molecular
2019;69:137–58. https://doi.org/10.1016/j.preteyeres.2018.10.004. and functional characterization of elovl4 genes in Sparus aurata and Solea
[69] Agaba M, Tocher DR, Dickson CA, Dick JR, Teale AJ. Zebrafish cDNA encoding senegalensis pointing to a critical role in very long-chain (>C24) fatty acid
multifunctional fatty acid elongase involved in production of eicosapentaenoic synthesis during early neural development of fish. Int J Mol Sci 2020;21:3514.
(20:5n-3) and docosahexaenoic (22:6n-3) acids. Mar Biotechnol 2004;6:251–61. https://doi.org/10.3390/ijms21103514.
https://doi.org/10.1007/s10126-003-0029-1. [91] Jin M, Monroig Ó, Navarro JC, Tocher DR, Zhou Q. Molecular and functional
[70] Agaba MK, Tocher DR, Zheng X, Dickson CA, Dick JR, Teale AJ. Cloning and characterisation of two elovl4 elongases involved in the biosynthesis of very long-
functional characterisation of polyunsaturated fatty acid elongases of marine and chain (> C24) polyunsaturated fatty acids in black seabream Acanthopagrus
freshwater teleost fish. Comp Biochem Physiol B 2005;142:342–52. https://doi. schlegelii. Comp Biochem Physiol B 2017;212:41–50. https://doi.org/10.1016/j.
org/10.1016/j.cbpb.2005.08.005. cbpb.2017.06.008.
[71] Oboh A. Investigating the long-chain polyunsaturated fatty acid biosynthesis of [92] Li S, Monroig Ó, Navarro JC, Yuan Y, Xu W, Mai K, et al. Molecular cloning and
the African catfish Clarias gariepinus (Burchell, 1822). PhD Thesis. University of functional characterization of a putative Elovl4 gene and its expression in
Stirling; 2018. http://hdl.handle.net/1893/27282. response to dietary fatty acid profiles in orange-spotted grouper Epinephelus
[72] Tinti E, Geay F, Rodrigues ML, Kestemont P, Perpète EA, Michaux C. Molecular coioide. Aquac Res 2017;48:537–52. https://doi.org/10.1111/are.12902.
cloning and 3D model of a fatty-acid elongase in a carnivorous freshwater teleost, [93] Zhu K, Song L, Guo H, Guo L, Zhang N, Liu B, et al. Elovl4a participates in LC-
the European perch (Perca fluviatilis). 3 Biotech 2019;9:242. https://doi.org/ PUFA biosynthesis and is regulated by PPARαb in golden pompano Trachinotus
10.1007/s13205-019-1773-x. ovatus (Linnaeus 1758). Sci Rep UK 2019;9:4684. https://doi.org/10.1038/
[73] Carmona-Antoñanzas G, Tocher DR, Taggart JB, Leaver MJ. An evolutionary s41598-019-41288-w.
perspective on Elovl5 fatty acid elongase: comparison of Northern pike and [94] Monroig Ó, Rotllant J, Sanchez E, Cerda-Reverter JM, Tocher DR. Expression of
duplicated paralogs from Atlantic salmon. BMC Evol Biol 2013;13:85. https:// long-chain polyunsaturated fatty acid (LC-PUFA) biosynthesis genes during
doi.org/10.1186/1471-2148-13-85. zebrafish Danio rerio early embryogenesis. BBA Mol Cell Biol L 2009;1791:
[74] Gregory MK, James MJ. Rainbow trout (Oncorhynchus mykiss) Elovl5 and Elovl2 1093–101. https://doi.org/10.1016/j.bbalip.2009.07.002.
differ in selectivity for elongation of omega-3 docosapentaenoic acid. BBA Mol [95] Xu W, Wang S, You C, Zhang Y, Monroig Ó, Tocher DR, et al. The catadromous
Cell Biol L 2014:1656–60. https://doi.org/10.1016/j.bbalip.2014.10.001. teleost Anguilla japonica has a complete enzymatic repertoire for the biosynthesis
[75] Morais S, Monroig Ó, Zheng X, Leaver MJ, Tocher DR. Highly unsaturated fatty of docosahexaenoic acid from α-linolenic acid: cloning and functional
acid synthesis in Atlantic salmon: characterization of ELOVL5-and ELOVL2-like characterization of an Elovl2 elongase. Comp Biochem Physiol B 2020;240:
elongases. Mar Biotechnol 2009;11:627–39. https://doi.org/10.1007/s10126- 110373. https://doi.org/10.1016/j.cbpb.2019.110373.
009-9179-0. [96] Machado AM, Tørresen OK, Kabeya N, Couto A, Petersen B, Felício M, et al. “Out
[76] Kim SH, Kim SY, Kim EK, Roh KH, Kim JB, Kim KS, et al. Identification and of the can”: a draft genome assembly, liver transcriptome, and nutrigenomics of
functional characterization of polyunsaturated fatty acid elongase (McELOVL5) the European sardine, Sardina pilchardus. Genes 2018;9:485. https://doi.org/
gene from pike eel (Muraenesox cinereus). Biotechnol Lett 2014;36:29–37. https:// 10.3390/genes9100485.
doi.org/10.1007/s10529-013-1344-x. [97] Li Y, Wen Z, You C, Xie Z, Tocher DR, Zhang Y, et al. Genome wide identification
and functional characterization of two LC-PUFA biosynthesis elongase (elovl8)

13
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

genes in rabbitfish (Siganus canaliculatus). Aquaculture 2020;522:735127. striata and Danio rerio elovl5 elongase. Fish Physiol Biochem 2020;46:1349–59.
https://doi.org/10.1016/j.aquaculture.2020.735127. https://doi.org/10.1007/s10695-020-00793-w.
[98] Bhandari S, Lee JN, Kim YI, Nam IK, Kim SJ, Kim SJ, et al. The fatty acid chain [120] Dong Y, Wang S, You C, Xie D, Jiang Q, Li Y. Hepatocyte nuclear factor 4α
elongase, Elovl1, is required for kidney and swim bladder development during (Hnf4α) is involved in transcriptional regulation of Δ6/Δ5 fatty acyl desaturase
zebrafish embryogenesis. Organogenesis 2016;12:78–93. https://doi.org/ (Fad) gene expression in marine teleost Siganus canaliculatus. Comp Biochem
10.1080/15476278.2016.1172164. Physiol B 2019;239:110353. https://doi.org/10.1016/j.cbpb.2019.110353.
[99] Ohno Y, Suto S, Yamanaka M, Mizutani Y, Mitsutake S, Igarashi Y, et al. ELOVL1 [121] Li Y, Yin Z, Dong Y, Wang S, Monroig Ó, Tocher DR, et al. Pparγ is involved in the
production of C24 acyl-CoAs is linked to C24 sphingolipid synthesis. Proc Natl transcriptional regulation of liver LC-PUFA biosynthesis by targeting the Δ6Δ5
Acad Sci USA 2010;107:18439–44. https://doi.org/10.1073/pnas.1005572107. fatty acyl desaturase gene in the marine teleost Siganus canaliculatus. Mar
[100] Chen J, Cui Y, Yan J, Jiang J, Cao X, Gao J. Molecular characterization of Biotechnol 2019;21:19–29. https://doi.org/10.1007/s10126-018-9854-0.
elongase of very long-chain fatty acids 6 (elovl6) genes in Misgurnus [122] Li Y, Zeng X, Dong Y, Chen C, You C, Tang G, et al. Hnf4α is involved in LC-PUFA
anguillicaudatus and their potential roles in adaptation to cold temperature. Gene biosynthesis by up-regulating gene transcription of elongase in marine teleost
2018;666:134–44. https://doi.org/10.1016/j.gene.2018.05.019. Siganus canaliculatus. Int J Mol Sci 2018;19:3193. https://doi.org/10.3390/
[101] Li Y, Pang Y, Xiang X, Du J, Mai K, Ai Q. Molecular cloning, characterization, and ijms19103193.
nutritional regulation of Elovl6 in large yellow croaker (Larimichthys crocea). Int J [123] Hayhurst GP, Lee YH, Lambert G, Ward JM, Gonzalez FJ. Hepatocyte nuclear
Mol Sci 2019;20:1801. https://doi.org/10.3390/ijms20071801. factor 4 alpha (nuclear receptor 2A1) is essential for maintenance of hepatic gene
[102] Li Y, Pang Y, Zhao Z, Xiang X, Ma K, Ai Q. Molecular characterization, nutritional expression and lipid homeostasis. Mol Cell Biol 2001;21:1393–403. https://doi.
and insulin regulation of Elovl6 in rainbow trout (Oncorhynchus mykiss). org/10.1128/MCB.21.4.1393-1403.2001.
Biomolecules 2020;10:264. https://doi.org/10.3390/biom10020264. [124] Leaver MJ, Taggart JB, Villeneuve L, Bron JE, Guy DR, Bishop SC, et al.
[103] Mah MQ, Kuah MK, Ting SY, Merosha P, Janaranjani M, Goh PT, et al. Molecular Heritability and mechanisms of n-3 long chain polyunsaturated fatty acid
cloning, phylogenetic analysis and functional characterisation of an Elovl7-like deposition in the flesh of Atlantic salmon. Comp Biochem Physiol D 2011;6:62–9.
elongase from a marine crustacean, the orange mud crab (Scylla olivacea). Comp https://doi.org/10.1016/j.cbd.2010.04.002.
Biochem Physiol B 2019;232:60–71. https://doi.org/10.1016/j. [125] Xu Y, Luo Z, Wu K, Fan Y, You W, Zhang L. Structure and functional analysis of
cbpb.2019.01.011. promoters from two liver isoforms of CPT I in grass carp Ctenopharyngodon Idella.
[104] Nara TY, He WS, Tang C, Clarke SD, Nakamura MT. The E-box like sterol Int J Mol Sci 2017;18:2405. https://doi.org/10.3390/ijms18112405.
regulatory element mediates the suppression of human Δ6 desaturase gene by [126] Wang S, Chen J, Jiang D, Zhang Q, You C, Tocher DR, et al. Hnf4α is involved in
highly unsaturated fatty acids. Biochem Bioph Res Co 2002;296:111–7. https:// the regulation of vertebrate LC-PUFA biosynthesis: insights into the regulatory
doi.org/10.1016/S0006-291X(02)00851-3. role of Hnf4α on expression of liver fatty acyl desaturases in the marine teleost
[105] Qin Y, Dalen KT, Gustafsson JA, Nebb HI. Regulation of hepatic fatty acid Siganus canaliculatus. Fish Physiol Biochem 2018;44:805–15. https://doi.org/
elongase 5 by LXRα-SREBP-1c. BBA Mol Cell Biol L 2009;1791:140–7. https://doi. 10.1007/s10695-018-0470-8.
org/10.1016/j.bbalip.2008.12.003. [127] Goldstein JL, DeBose-Boyd RA, Brown MS. Protein sensors for membrane sterols.
[106] Tang C, Cho HP, Nakamura MT, Clarke SD. Regulation of human Δ6 desaturase Cell 2006;124:35–46. https://doi.org/10.1016/j.cell.2005.12.022.
gene transcription: identification of a functional direct repeat-1 element. J Lipid [128] Wójcicka G, Jamroz-Wiśniewska A, Horoszewicz K, Bełtowski J. Liver X receptors
Res 2003;44:686–95. https://doi.org/10.1194/jlr.M200195-JLR200. (LXRs). Part I: structure, function, regulation of activity, and role in lipid
[107] Wahl HG, Kausch C, Machicao F, Rett K, Stumvoll M, Haring HU. Troglitazone metabolism. Postȩp Hig Med Dośw 2007;61:736–59. https://pubmed.ncbi.nlm.
downregulates Δ6 desaturase gene expression in human skeletal muscle cell nih.gov/18063918/.
cultures. Diabetes 2002;51:1060–5. https://doi.org/10.2337/diabetes.51.4.1060. [129] Cruz-Garcia L, Minghetti M, Navarro I, Tocher DR. Molecular cloning, tissue
[108] Varin A, Thomas C, Ishibashi M, Ménégaut L, Gautier T, Trousson A, et al. Liver X expression and regulation of liver X receptor (LXR) transcription factors of
receptor activation promotes polyunsaturated fatty acid synthesis in Atlantic salmon (Salmo salar) and rainbow trout (Oncorhynchus mykiss). Comp
macrophages: relevance in the context of atherosclerosis. Arterioscl Throm Vas Biochem Physiol B 2009;153:81–8. https://doi.org/10.1016/j.cbpb.2009.02.001.
2015;35:1357–65. https://doi.org/10.1161/atvbaha.115.305539. [130] Minghetti M, Leaver MJ, Tocher DR. Transcriptional control mechanisms of genes
[109] Zheng X, Leaver M, Tocher DR. Long-chain polyunsaturated fatty acid synthesis in of lipid and fatty acid metabolism in the Atlantic salmon (Salmo salar L.)
fish: comparative analysis of Atlantic salmon (Salmo salar L.) and Atlantic cod established cell line, SHK-1. BBA Mol Cell Biol 2011;L 1811:194–202. https://doi.
(Gadus morhua L.) delta 6 fatty acyl desaturase gene promoters. Comp Biochem org/10.1016/j.bbalip.2010.12.008.
Physiol B 2009;154:255–63. https://doi.org/10.1016/j.cbpb.2009.06.010. [131] Dong X, Xu H, Mai K, Xu W, Zhang Y, Ai Q. Cloning and characterization of
[110] Geay F, Zambonino-Infante J, Reinhardt R, Kuhl H, Santigosa E, Cahu C, et al. SREBP-1 and PPAR-α in Japanese seabass Lateolabrax japonicus, and their gene
Characteristics of Fads2 gene expression and putative promoter in European sea expressions in response to different dietary fatty acid profiles. Comp Biochem
bass (Dicentrarchus labrax): comparison with salmonid species and analysis of CpG Physiol B 2015;180:48–56. https://doi.org/10.1016/j.cbpb.2014.10.001.
methylation. Mar Genom 2012;5:7–13. https://doi.org/10.1016/j. [132] Zhang Q, You C, Liu F, Zhu W, Wang S, Xie D, et al. Cloning and characterization
margen.2011.08.003. of Lxr and Srebp1, and their potential roles in regulation of LC-PUFA biosynthesis
[111] Xu H, Dong X, Ai Q, Mai K, Xu W, Zhang Y, et al. Regulation of tissue LC-PUFA in rabbitfish Siganus canaliculatus. Lipids 2016;51:1–13. https://doi.org/10.1007/
contents, Delta6 fatty acyl desaturase (FADS2) gene expression and the s11745-016-4176-3.
methylation of the putative FADS2 gene promoter by different dietary fatty acid [133] Carmona-Antoñanzas G, Tocher DR, Martinez-Rubio L, Leaver MJ. Conservation
profiles in Japanese seabass (Lateolabrax japonicus). PloS One 2014;9:e87726. of lipid metabolic gene transcriptional regulatory networks in fish and mammals.
https://doi.org/10.1371/journal.pone.0087726. Gene 2014;534:1–9. https://doi.org/10.1016/j.gene.2013.10.040.
[112] Dong X, Tan P, Cai Z, Xu H, Li J, Ren W, et al. Regulation of FADS2 transcription [134] Berger J, Moller DE. The mechanisms of action of PPARs. Annu Rev Med 2002;53:
by SREBP-1 and PPAR-α influences LC-PUFA biosynthesis in fish. Sci Rep 2017;7: 409–35. https://doi.org/10.1146/annurev.med.53.082901.104018.
40024. https://doi.org/10.1038/srep40024. [135] Willson TM, Brown PJ, Sternbach DD, Henke BR. The PPARs: from orphan
[113] Xie D, Fu Z, Wang S, You C, Monroig Ó, Tocher DR, et al. Characteristics of the receptors to drug discovery. J Med Chem 2000;43:527–50. https://doi.org/
Fads2 gene promoter in marine teleost Epinephelus coioides and role of Sp1- 10.1021/jm990554g.
binding site in determining promoter activity. Sci Rep 2018;8:5305. https://doi. [136] Turner J, Crossley M. Mammalian krüppel-like transcription factors: more than
org/10.1038/s41598-018-23668-w. just a pretty finger. Trends Biochem Sci 1999;24:236–40. https://doi.org/
[114] Dong Y, Zhao J, Chen J, Wang S, Liu Y, Zhang Q, et al. Cloning and 10.1016/S0968-0004(99)01406-1.
characterization of ∆6/∆5 fatty acyl desaturase (Fad) gene promoter in the marine [137] Samson S. Role of Sp1 in insulin regulation of gene expression. J Mol Endocrinol
teleost Siganus canaliculatus. Gene 2018;647:174–80. https://doi.org/10.1016/j. 2002;29:265–79. https://doi.org/10.1677/jme.0.0290265.
gene.2018.01.031. [138] Reed BD, Charos AE, Szekely AM, Weissman SM, Snyder M. Genome-wide
[115] Tay SS, Kuah MK, Shu-Chien AC. Transcriptional activation of zebrafish Fads2 occupancy of SREBP1 and its partners NFY and SP1 reveals novel functional roles
promoter and its transient transgene expression in yolk syncytial layer of and combinatorial regulation of distinct classes of genes. PLoS Genet 2008;4:
zebrafish embryos. Sci Rep 2018;8:3874. https://doi.org/10.1038/s41598-018- e1000133. https://doi.org/10.1371/journal.pgen.1000133.
22157-4. [139] Bennett MK, Osborne TF. Nutrient regulation of gene expression by the sterol
[116] Dong Y, Wang S, Chen J, Zhang Q, Liu Y, You C, et al. Hepatocyte nuclear factor regulatory element binding proteins: increased recruitment of gene-specific
4α (HNF4α) is a transcription factor of vertebrate fatty acyl desaturase gene as coregulatory factors and selective hyperacetylation of histone H3 in vivo. Proc
identified in marine teleost Siganus canaliculatus. PloS One 2016;11:e0160361. Natl Acad Sci USA 2000;97:6340–4. https://doi.org/10.1073/pnas.97.12.6340.
https://doi.org/10.1371/journal.pone.0160361. [140] Li Y, Hu C, Zheng Y, Xia X, Xu W, Wang S, et al. The effects of dietary fatty acids
[117] Carmona-Antoñanzas G, Zhen X, Tocher DR, Leaver MJ. Regulatory divergence of on liver fatty acid composition and Δ6-desaturase expression differ with ambient
homologous Atlantic salmon elovl5 genes following the salmonid-specific whole- salinities in Siganus canaliculatus. Comp Biochem Physiol B 2008;151:183–90.
genome duplication. Gene 2016;591:34–42. https://doi.org/10.1016/j. https://doi.org/10.1016/j.cbpb.2008.06.013.
gene.2016.06.056. [141] Xie D, Wang S, You C, Chen F, Tocher DR, Li Y. Characteristics of LC-PUFA
[118] Li Y, Zhao J, Dong Y, Yin Z, Li Y, Liu Y, et al. Sp1 is involved in vertebrate LC- biosynthesis in marine herbivorous teleost Siganus canaliculatus under different
PUFA biosynthesis by upregulating the expression of liver desaturase and ambient salinities. Aquacult Nutr 2015;21:541–51. https://doi.org/10.1111/
elongase genes. Int J Mol Sci 2019;20:5066. https://doi.org/10.3390/ anu.12178.
ijms20205066. [142] Liu Y, Zhang Q, Dong Y, You C, Wang S, Li Y, et al. Establishment of a hepatocyte
[119] Goh PT, Kuah MK, Chew YS, Teh HY, Shu-Chien AC. The requirements for sterol line for studying biosynthesis of long-chain polyunsaturated fatty acids from a
regulatory element-binding protein (Srebp) and stimulatory protein 1 (Sp1)- marine teleost, the white-spotted spinefoot Siganus canaliculatus. J Fish Biol 2017;
binding elements in the transcriptional activation of two freshwater fish Channa 91:603–16. https://doi.org/10.1111/jfb.13375.

14
D. Xie et al. Progress in Lipid Research 82 (2021) 101095

[143] Sun J, Zheng L, Chen C, Zhang J, You C, Zhang Q, et al. MicroRNAs involved in [158] Szabo G, Bala S. MicroRNAs in liver diseases. Nat Rev Gastroenterol Hepatol
the regulation of LC-PUFA biosynthesis in teleosts: miR-33 enhances LC-PUFA 2013;10:542–52. https://doi.org/10.1038/nrgastro.2013.87.
biosynthesis in Siganus canaliculatus by targeting insig1 which in turn up-regulates [159] Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs.
srebp1. Mar Biotechnol 2019;21:475–87. https://doi.org/10.1007/s10126-019- Cell 2009;136:642–55. https://doi.org/10.1016/j.cell.2009.01.035.
09895-w. [160] Flowers E, Froelicher ES, Aouizerat BE. MicroRNA regulation of lipid metabolism.
[144] Janani C, Kumari BR. PPAR gamma gene–a review. Diabetes Metab Syndr 2015;9: Metabolism 2013;62:12–20. https://doi.org/10.1016/j.metabol.2012.04.009.
46–50. https://doi.org/10.1016/j.dsx.2014.09.015. [161] Zhang Q, Xie D, Wang S, You C, Monroig Ó, Tocher DR, et al. miR-17 is involved
[145] You C, Jiang D, Zhang Q, Xie D, Wang S, Dong Y, et al. Cloning and expression in the regulation of LC-PUFA biosynthesis in vertebrates: effects on liver
characterization of peroxisome proliferator-activated receptors (PPARs) with expression of a fatty acyl desaturase in the marine teleost Siganus canaliculatus.
their agonists, dietary lipids, and ambient salinity in rabbitfish Siganus BBA Mol Cell Biol L 2014;1841:934–43. https://doi.org/10.1016/j.
canaliculatus. Comp Biochem Physiol B 2017;206:54–64. https://doi.org/ bbalip.2014.03.009.
10.1016/j.cbpb.2017.01.005. [162] Zhang Q, You C, Wang S, Dong Y, Monroig O, Tocher DR, et al. The miR-33 gene
[146] Zuo R, Mai K, Xu W, Dong X, Ai Q. Molecular cloning, tissue distribution and is identified in a marine teleost: a potential role in regulation of LC-PUFA
nutritional regulation of a Δ6-fatty acyl desaturase-like enzyme in large yellow biosynthesis in Siganus canaliculatus. Sci Rep 2016;6:32909–19. https://doi.org/
croaker (Larimichthys crocea). Aquac Res 2016;47:445–59. https://doi.org/ 10.1038/srep32909.
10.1111/are.12505. [163] Chen C, Zhang J, Zhang M, You C, Liu Y, Wang S, et al. miR-146a is involved in
[147] Wang S, Wang M, Zhang H, Yan X, Guo H, You C, et al. Long-chain the regulation of vertebrate LC-PUFA biosynthesis by targeting elovl5 as
polyunsaturated fatty acid metabolism in carnivorous marine teleosts: Insight into demonstrated in rabbitfish Siganus canaliculatus. Gene 2018;676:306–14. https://
the profile of endogenous biosynthesis in golden pompano Trachinotus ovatus. doi.org/10.1016/j.gene.2018.08.063.
Aquac Res 2020;51:623–35. https://doi.org/10.1111/are.14410. [164] Chen C, Wang S, Zhang M, Chen B, You C, Xie D, et al. miR-24 is involved in
[148] Seiliez I, Panserat S, Corraze G, Kaushik S, Bergot P. Cloning and nutritional vertebrate LC-PUFA biosynthesis as demonstrated in marine teleost Siganus
regulation of a delta6-desaturase-like enzyme in the marine teleost gilthead canaliculatus. BBA Mol Cell Biol L 2019;1864:619–28. https://doi.org/10.1016/j.
seabream (Sparus aurata). Comp Biochem Physiol B 2003;135:449–60. https:// bbalip.2019.01.010.
doi.org/10.1016/S1096-4959(03)00111-8. [165] Chen C, Wang S, Hu Y, Zhang M, He X, You C, et al. miR-26a mediates LC-PUFA
[149] Izquierdo MS, Robaina L, Juárez-Carrillo E, Oliva V, Hernández-Cruz CM, biosynthesis by targeting the Lxrα-Srebp1 pathway in the marine teleost Siganus
Afonso JM. Regulation of growth, fatty acid composition and delta 6 desaturase canaliculatus. J Biol Chem 2020;295:13875–86. https://doi.org/10.1074/jbc.
expression by dietary lipids in gilthead seabream larvae (Sparus aurata). Fish RA120.014858.
Physiol Biochem 2008;34:117–27. https://doi.org/10.1007/s10695-007-9152-7. [166] Chen C, Zhang M, Li Y, Wang S, Xie D, Wen X, et al. Identification of miR-145 as a
[150] Egea M, Metón I, Córdoba M, Fernández F, Baanante IV. Role of Sp1 and SREBP- key regulator involved in LC-PUFA biosynthesis by targeting hnf4α in the marine
1a in the insulin-mediated regulation of glucokinase transcription in the liver of teleost Siganus canaliculatus. J Agri Food Chem 2020;68:15123–33. https://doi.
gilthead sea bream (Sparus aurata). Gen Comp Endocr 2008;155:359–67. https:// org/10.1021/acs.jafc.0c04649.
doi.org/10.1016/j.ygcen.2007.06.018. [167] Sun J, Chen C, You C, Liu Y, Ma H, Monroig Ó, et al. The miR-15/16 cluster is
[151] Cocci P, Mosconi G, Arukwe A, Mozzicafreddo M, Angeletti M, Aretusi G, et al. involved in the regulation of vertebrate LC-PUFA biosynthesis by targeting pparγ
Effects of diisodecyl phthalate on PPAR: RXR-dependent gene expression as demonstrated in rabbitfish Siganus canaliculatus. Mar Biotechnol 2020;22:
pathways in sea bream hepatocytes. Chem Res Toxicol 2015;28:935–47. https:// 475–87. https://doi.org/10.1007/s10126-020-09969-0.
doi.org/10.1021/tx500529x. [168] Horie T, Nishino T, Baba O, Kuwabara Y, Nakao T, Nishiga M, et al. MicroRNA-33
[152] Cocci P, Mozzicafreddo M, Angeletti M, Mosconi G, Palermo FA. Differential regulates sterol regulatory element-binding protein 1 expression in mice. Nat
tissue regulation of peroxisome proliferator-activated receptor α (PPARα) and Commun 2013;4:2883. https://doi.org/10.1038/ncomms3883.
cannabinoid receptor 1 (CB1) gene transcription pathways by diethylene glycol [169] Radhakrishnan A, Ikeda Y, Kwon HJ, Brown MS, Goldstein JL. Sterol-regulated
dibenzoate (DEGB): preliminary observations in a seabream (Sparus aurata) in transport of SREBPs from endoplasmic reticulum to Golgi: Oxysterols block
vivo model. Environ Toxicol Phar 2017;55:87–93. https://doi.org/10.1016/j. transport by binding to Insig. Proc Natl Acad Sci USA 2007;104:6511–8. https://
etap.2017.08.015. doi.org/10.1073/pnas.0700899104.
[153] Betancor MB, Sprague M, Montero D, Usher S, Sayanova O, Campbell P, et al. [170] Poy MN, Eliasson L, Krutzfeldt J, Kuwajima J, Ma X, Macdonald PE, et al.
Replacement of marine fish oil with de novo omega-3 oils from transgenic A pancreatic islet-specific microRNA regulates insulin secretion. Nature 2004;
Camelina sativa in feeds for gilthead sea bream (Sparus aurata L.). Lipids 2016;51: 432:226–30. https://doi.org/10.1038/nature03076.
1171–91. https://doi.org/10.1007/s11745-016-4191-4. [171] Yu J, Wang F, Yang GH, Wang FL, Ma YN, Du ZW, et al. Human microRNA
[154] Houston SJ, Karalazos V, Tinsley J, Betancor MB, Martin SA, Tocher DR, et al. The clusters: genomic organization and expression profile in leukemia cell lines.
compositional and metabolic responses of gilthead seabream (Sparus aurata) to a Biochem Bioph Res Co 2006;349:59–68. https://doi.org/10.1016/j.
gradient of dietary fish oil and associated n-3 long-chain PUFA content. Brit J bbrc.2006.07.207.
Nutr 2017;118:1010–22. https://doi.org/10.1017/S0007114517002975. [172] Fan Y, Arbab AAI, Zhang H, Yang Y, Lu X, Han Z, et al. MicroRNA-193a-5p
[155] Perera E, Turkmen S, Simó-Mirabet P, Zamorano MJ, Xu H, Naya-Català F, et al. regulates the synthesis of polyunsaturated fatty acids by targeting fatty acid
Stearoyl-CoA desaturase (scd1a) is epigenetically regulated by broodstock desaturase 1 (FADS1) in bovine mammary epithelial cells. Biomolecules 2021;11:
nutrition in gilthead sea bream (Sparus aurata). Epigenetics 2020;15:536–53. 157. https://doi.org/10.3390/biom11020157.
https://doi.org/10.1080/15592294.2019.1699982. [173] Zhang HM, Kuang S, Xiong X, Gao T, Liu C, Guo AY. Transcription factor and
[156] Turkmen S, Perera E, Zamorano MJ, Simo-Mirabet P, Xu H, Perez-Sanchez J, et al. microRNA co-regulatory loops: important regulatory motifs in biological
Effects of dietary lipid composition and fatty acid desaturase 2 expression in processes and diseases. Brief Bioinform. 2015;16:45–58. https://doi.org/
broodstock gilthead seabream on lipid metabolism-related genes and methylation 10.1093/bib/bbt085.
of the fads2 gene promoter in their offspring. Int J Mol Sci 2019;20:6250. https:// [174] Tocher DR, Betancor MB, Sprague M, Olsen RE, Napier JA. Omega-3 long-chain
doi.org/10.3390/ijms20246250. polyunsaturated fatty acids, EPA and DHA: Bridging the gap between supply and
[157] Nachtergaele S, He C. The emerging biology of RNA post-transcriptional demand. Nutrients 2019;11:89. https://doi.org/10.3390/nu11010089.
modifications. RNA Biol 2016;14:156–63. https://doi.org/10.1080/
15476286.2016.1267096.

15

You might also like