an alternative to fish oils metabolic engineering of oil-seed crops to produce

Upload: alcidan

Post on 03-Jun-2018

219 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    1/12

    Review

    An alternative to fish oils: Metabolic engineering of oil-seed crops to produceomega-3 long chain polyunsaturated fatty acids

    Mnica Venegas-Calern a,b, Olga Sayanova a, Johnathan A. Napier a,*

    a Department of Biological Chemistry, Rothamsted Research, Harpenden, Herts AL5 2JQ, UKb Instituto de la Grasa, CSIC, Av. Padre Garcia Tejero 4, E-41012 Seville, Spain

    a r t i c l e i n f o

    Article history:

    Received 17 September 2009

    Received in revised form 13 October 2009

    Accepted 20 October 2009

    Keywords:

    Polyunsaturated fatty acids

    Plants

    Omega-3 fatty acids

    Desaturases

    Elongases

    Transgenic plant

    a b s t r a c t

    It is now accepted that omega-3 polyunsaturated fatty acids, especially eicosapentaenoic acid (EPA;

    20:5D5,8,11,14,17) and docosahexaenoic acid (DHA, 22:6D4,7,10,13,16,19) play important roles in a

    number of aspects of human health, with marine fish rich in these beneficial fatty acids our primary die-

    tary source. However, over-fishing and concerns about pollution of the marine environment indicate a

    need to develop alternative, sustainable sources of very long chain polyunsaturated fatty acids (VLC-

    PUFAs) such as EPAand DHA. A number of different strategies have been considered, with one of the most

    promising being transgenic plants reverse-engineered to produce these so-called fish oils. Considerable

    progress has been made towards this goal and in this review we will outline the recent achievements in

    demonstrating the production of omega-3 VLC-PUFAs in transgenic plants. We will also consider how

    these enriched oils will allow the development of nutritionally-enhanced food products, suitable either

    for direct human ingestion or for use as an animal feedstuff. In particular, the requirements of aquacul-

    ture for omega-3 VLC-PUFAs will act as a strong driver for the development of such products. In addition,

    biotechnological research on the synthesis of VLC-PUFAs has provided new insights into the complexities

    of acyl-channelling and triacylglycerol biosynthesis in higher plants.

    2009 Elsevier Ltd. All rights reserved.

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109

    2. Omega-3 long chain polyunsaturated fatty acids (x3 LC-PUFAs) in humans health. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1093. Characterization of VLC-PUFAs biosynthetic pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110

    4. Metabolic engineering to produce VLC-PUFAs in higher plants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112

    5. Crucial issues: optimization the levels of LC-PUFA in transgenic plants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115

    5.1. The identification of superior desaturases. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115

    5.2. Identification of a VLC-PUFA-specific acyl-exchange mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115

    5.3. Maintenance of a continuous flux of substrates through the VLC-PUFA biosynthetic pathway without significant loss to TAG . . . . . . . 115

    5.4. Optimizing the fatty acid elongase. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116

    5.5. Modulating the acyl-CoA pool . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116

    5.6. Co-ordinated expression of transgenes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116

    5.7. Appropriate localisation of transgene-derived activities. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1166. Conclusions and future perspectives. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116

    Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117

    0163-7827/$ - see front matter 2009 Elsevier Ltd. All rights reserved.doi:10.1016/j.plipres.2009.10.001

    Abbreviations: ALA, a-linolenic acid; ARA, arachidonic acid; DAG, diacylglycerol; DGAT, diacylgylcerol acyltransferase; DHA, docosahexaenoic acid; ECR, enoyl-CoAreductase; EFA, essential fatty acid; EPA, eicosapentaenoic acid; GLA, c-linolenic acid; HCD, hydroxyacyl-CoA dehydratase; KCS, ketoacyl-CoA synthase; KCR, b-ketoacyl-CoAreductase; LA, linoleic acid; LPCAT, acyl-CoA: lyso-phosphatidylcholine acyltransferase; PDAT, phospholipid: diacylglycerol acyltransferase; SDA, stearidonic acid; TAG,

    triacylglycerol; VLC-PUFA, very long chain polyunsaturated fatty acid.

    * Corresponding author. Tel.: +44 (0) 1582 763133; fax: +44 (0) 1582 763010.

    E-mail address: [email protected](J.A. Napier).

    Progress in Lipid Research 49 (2010) 108119

    Contents lists available at ScienceDirect

    Progress in Lipid Research

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / p l i p r e s

    http://dx.doi.org/10.1016/j.plipres.2009.10.001mailto:[email protected]://www.sciencedirect.com/science/journal/01637827http://www.elsevier.com/locate/plipreshttp://www.elsevier.com/locate/plipreshttp://www.sciencedirect.com/science/journal/01637827mailto:[email protected]://dx.doi.org/10.1016/j.plipres.2009.10.001
  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    2/12

    1. Introduction

    Very long chain polyunsaturated fatty acids (VLC-PUFAs) are

    fatty acids of 20 carbons or more in length with three or more

    methylene-interrupted double bonds in the cis position. These

    fatty acids can be grouped into two main families, omega-6 (or

    n-6) and omega-3 (or n-3) families, depending on the position of

    the first double bond proximal to the methyl end of the fatty acid.

    VLCLC-PUFAs are vital constituents of human metabolism. In par-

    ticular, there is plentiful evidence (from epidemiology and dietary

    intervention studies) for the health-beneficial properties to hu-

    mans of dietary consumption of omega-3 VLC-PUFAs such as eico-

    sapentaenoic acid (EPA; 20:5D5,8,11,14,17) and docosahexaenoic

    acid (DHA; 22:6D4,7,10,13,16,19) [14]. This dietary requirement

    is almost certainly due to the fact that humans (like most animals)

    have a very limited capacity to synthesize these fatty acids from

    the essential precursor a-linolenic acid (ALA; 18:3D9,12,15) [5];therefore dietary intake of these fatty acids is a key aspect of hu-

    man nutrition[6]. The main source of EPA and DHA in the human

    diet is through the direct consumption of cold water marine fish

    (such as salmon, tuna, mackerel, and sardines) [6,7]. However,

    marine fish (like other animals) do not efficiently metabolise ALA

    to VLC-PUFAs, but accumulate them as a result of their dietary

    acquisition (though it should be noted that freshwater fish appear

    to have a greater capacity to synthesize EPA and DHA from ALA)

    [7,8]. The primary de novo synthesis sources of VLC-PUFAs are mar-

    ine microbes such as algae which form the base of an aquatic food

    web that culminates in the accumulation of these fatty acids in the

    lipids of the fish [9]. In these microorganisms, EPA and DHA are

    synthesized de novoby one of the two classes of biochemical path-

    way (reviewed on the text below). In addition, some fungi, mosses,

    bacteria and lower plants also have a capacity to synthesize signif-

    icant amounts of VLC-PUFAs [10]. In higher plants these fatty acids

    are almost completely absent, although plants are rich in the two

    essential dietary fatty acids linoleic acid (LA; 18:2D9,12) anda-lin-olenic acid (ALA; 18:3D9,12,15) that serve as the metabolic precur-

    sors for VLC-PUFA biosynthesis in animals[11].There is growing concern regarding the sustainability of global

    fish stocks (the predominant sources of omega-3 VLC-PUFA) be-

    cause marine fish stocks are in severe decline as a result of decades

    of over-fishing[12]. Moreover, environmental pollution of marine

    ecosystems has resulted in the accumulation of dioxins, heavy

    metals and polychlorinated biphenyls in fish, to the point of ques-

    tioning the benefits of fish consumption in human health [13]. Fi-

    nally, the expansion of industrialised aquaculture exacerbates the

    overexploitation of natural marine resources, since farmed fish re-

    quire omega-3 VLC-PUFA-containing feedstuffs. The marine oils

    used as aquaculture feedstocks are usually extracted from so-

    called trash species such as sand eels, which are specifically har-

    vested for this application (since they are not normally consumed

    by humans). However, the loss of these species from food-webs hasa profound impact on the overall stability of ecosystems [14].

    Aquaculture is certainly the largest consumer of fish-derived oils

    and currently even the most sophisticated husbandry of high value

    species such as salmon require the input of dietary fish oils to a le-

    vel significantly higher than that present in the finished product.

    Therefore aquaculture is (perhaps surprisingly, at least to the lay-

    person) a net consumer of fish oils and as such, not operating in

    a sustainable manner. In view of all of these points, there is a very

    obvious requirement for an alternative and sustainable source of

    VLC-PUFA for their use in human nutrition [1517].

    Perhaps the most obvious alternative to fish oils is via contained

    culture of the aquatic microbes which synthesize EPA and/or DHA.

    Approaches using microbiological sources to synthesize VLC-PUFA

    have been developed and are economically viable for specific highvalue applications (such as infant formula baby milk formula-

    tions [18]) in controlled culture systems. However, such systems

    are expensive to maintain and have limited flexibility for signifi-

    cant scale-up and requiring the appropriate microbiological facili-

    ties (such as fermenters) [19]. It is noteworthy that such

    fermentation-based systems are also sensitive to disruptions of

    power-supplies and have a significant environmental footprint.

    In view of all these factors, there is an obvious need for an alter-

    native, sustainable source of these important fatty acids. Oneattractive option is the use of transgenic plants to synthesize these

    fatty acids. Because no higher plant oilseeds produce VLC-PUFAs

    such as EPA and DHA, they must be reverse-engineered (the dis-

    covery of technological principles through deconstruction and

    analysis of component parts)[20]with this biosynthetic capacity

    by the introduction of this metabolic pathway from a suitable

    microbial source [16,17,20]. During the last ten years, genes encod-

    ing the primary enzymes involved in biosynthesis of these fatty

    acids have been successfully isolated from a range of VLC-PUFA-

    synthesising organisms with a number of these being heterolo-

    gously expressed (singly or in combination) in oil-seed crops

    [21,22] the promise and the prospects of these new transgenic

    crops will be considered in this review.

    2. Omega-3 long chain polyunsaturated fatty acids

    (x3 LC-PUFAs) in humans health

    All animals have lost the capacity to synthesize VLC-PUFAS due

    to the genetic absence ofD12 and D15-desaturase activities and, as

    a consequence, cannot produce linolenic acid (LA; 18:2D9,12n-6)

    anda-linolenic acid (ALA; 18:3D9,12,15 n-3) respectively from theprecursor oleic acid (18:1D9) [23]. However, they do have a lim-

    ited ability to synthesize ARA and EPA from these two dietary-

    essential fatty acids (EFAs) LA and ALA through a series of desatu-

    ration and elongation reactions (Fig. 1) [24]. Most dietary LA and

    ALA are b-oxidized to provide energy and only a small portion of

    them are converted to VLC-PUFAs [25]. It is estimated that the %

    conversion of ALA to EPA is 510%, whereas conversion of ALAto DHA is

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    3/12

    are ubiquitous in higher plant and play a crucial role in the synthe-

    sis of membrane lipids for the support of photosynthesis and also

    the precursors for the oxylipin jasmonic acid required for male fer-

    tility [22]. The presence of these EFA desaturases has been ob-

    served in some lower animals, most notably Caenorhabditis

    elegans. Genetic analysis of the role of these enzymes in C. elegans

    indicates that they play a vital role in development [34]and such

    studies indicate the usefulness of this nematode in studies on the

    role of EFAs and VLC-PUFAs in multicellular organisms [35].

    VLC-PUFAs are functional components that can modulate mem-

    brane fluidity and permeability. As a consequence they play crucial

    roles in human metabolism, not only playing structural roles inphospholipid bilayers but also acting as precursors to bioactive

    molecules. For example, both omega-6 and omega-3 C20 fatty

    acids are precursors of the eicosanoids, oxygenated VLC-PUFA

    metabolites involved in the regulation of inflammation, plaque

    aggregation, and vasoconstriction/dilation. Both EPA and ARA serve

    as substrates for the common cyclooxygenase and lipoxygenase

    enzymes; while omega-6 ARA produces more potent inflamma-

    tory, pro-aggregatory and inmuno-active eicosanoids (series-2),

    eicosanoids derived from omega-3 fatty acids (series-1 and ser-

    ies-3) are anti-inflammatory and modulate plaque aggregation

    and immune-reactivity[36,37]. Unsurprisingly, research has dem-

    onstrated that there are considerable health benefits to be gained

    from having a diet rich in VLC-PUFA, and in particular EPA and

    DHA. For example, the VLC-PUFAs ARA and DHA play an importantrole in neonatal health and development[3840], in particular the

    acquisition of ocular vision and brain development: it is for this

    reason that both these fatty acids are recommended for inclusion

    in infant formula milks[18]. Clinical trials have demonstrated pro-

    tective roles for EPA and DHA in the prevention of cardiovascular

    disease and there is also emerging evidence of these VLC-PUFAs

    protecting against metabolic syndrome and related disease states,

    such as obesity and type-2 diabetes[31,41]. More recently, protec-

    tive effects have been clinically studied for cancer[42], atheroscle-

    rosis, cognitive impairment, and various mental illness, in

    particular depression[29], childhood and attention-deficit hyper-

    activity disorder (ADHD)[43,44]and dementia[45]. Finally, there

    are epidemiological studies which extend the beneficial effects ofomega-3 VLC-PUFA to the immune system (including diseased

    states such as rheumatoid arthritis)[46], the reproductive system,

    skin barrier function [47] and other exciting emerging roles such as

    inflammation-resolution[48].

    3. Characterization of VLC-PUFAs biosynthetic pathways

    Over the last decade, all the primary genes involved in VLC-PUFA

    biosynthesis have been identified from a range of different species,

    including animals, fungi, plants and aquatic organisms. These genes

    can be classifiedinto the twodistinct enzymatic reactions that cata-

    lyse the primary biosynthetic process. The first of these are the

    microsomal fatty acid desaturases, so-called front end PUFA

    desaturases which belong to the N-terminal cytochrome b5-fusionsuperfamily, firstly identified in 1997 by Sayanova et al. [49]. The

    18:2 9,12Linoleico acid (LA)

    18:3 9,12,15-Linolenic acid (ALA)

    3 Des

    15 Des18:1 9

    Oleico acid (OA)

    12 Des

    18:0

    Stearic acid (SA)

    9 Des

    Plants

    Animals

    Micobial 4 pathwayMammalian Sprecher

    pathway

    Conventional 6-pathway

    Alternative

    8-pathwayAlternative

    8-pathway

    Fig. 1. Aerobic VLC-PUFA biosynthetic pathways. The various routes for synthesis of arachidonic acid (ARA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) are

    shown, as mediated by the consecutive action of desaturases and elongases. The predominant D6-pathway is shown, as is the alternative D8-pathway. Two routes for DHAsynthesis are shown, microbial D4-pathway and mammalian Sprecher pathway. Des = desaturase, Elo = elongase.

    110 M. Venegas-Calern et al. / Progress in Lipid Research 49 (2010) 108119

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    4/12

    cytochrome b5 domain is assumed to be involved in the electron

    transport chain required for acyl-desaturation, and currently all

    known examples of microsomal VLC-PUFA desaturases contain this

    N-terminal extension. This is in contrast to theD12 and D15-desat-

    urases found in plants, algae and some fungi which lack any such

    cytochrome b5 domain. Another characteristic of the front end

    desaturases is thesubstitutionof histidine by glutamine in the third

    histidine box (consensus sequence QX[23]HH). The presence

    of diagnostic motifs such as a cytocromeb5 domain and this variant

    His box have greatly facilitated the identification of candidate

    front end desaturases from animals, fungi, and algae, and also

    from the few plant species (borage, evening primrose, black currant

    andEchium) that carry out D6-desaturationof LA and ALA [50]. One

    crucial observation regarding the microsomal desaturases from

    lower eukaryotes is that very many of these enzymes utilize glycer-

    olipid-linked substrates, in particular fatty acids esterified to the

    sn-2 position of glycerolipids. This is not the case in animals, where

    the substrates for these enzyme activities are generally believed to

    be acyl-CoAs[5153].

    The second key enzymatic reaction in the synthesis of VLC-

    PUFA is elongation, which also occurs in the ER. The fatty acid elon-

    gation reaction consists of four sequencial activities: condensation

    of the substrate fatty acid with malonyl-CoA (b-ketoacyl-CoA syn-

    thase; KCS), reduction (b-ketoacyl-CoA reductase; KCR), dehydra-

    tion (hydroxyacyl-CoA dehydratase; HCD), and a second

    reduction (enoyl-CoA reductase; ECR) [54] and resulting in a

    two-carbon chain elongation of the input substrate fatty acid.

    The condensing enzymes are considered to be rate-limiting and

    the regulators of substrate-specificity with regard to chain length

    and pattern of double bonds. Perhaps unexpectedly, the expression

    of sequences encoding b-ketoacyl-CoA synthase activities alone are

    able to reconstitute a heterologous elongating activity without

    requirement for the co-expression of any other components of

    the elongase[5557]. It is therefore for this reason that KCSs are

    often (semantically incorrectly) referred to as elongases. It is as-

    sumed that the ability of heterologously expressed KCSs to appar-

    ently direct the elongation of substrate fatty acids is due to theinteraction between endogenous core elongase components

    (KCR, HCD, ECD) and the exogenous KCS in the absence of any

    of these three latter components, elongation cannot occur. It

    should be noted that KCS condensing enzymes can be divided into

    two distinct groups. A first group comprises the so-called ELO-like

    sequences (named after the yeast ELO genes, which are required

    for the synthesis of saturated very long chain fatty acids found in

    sphingolipids [58]) some of which involved in VLC-PUFA biosyn-

    thesis, which have been cloned from a number of species including

    mammals, fungi (e.g. Mortierella alpina) [59], and aquatic algae (e.g.

    Isochrysis galbana)[60]. A second class of unrelated plant-specific

    KCS activities are known as FAE1-like enzymes (so-called after

    the founding member of this family, FAE1 fatty acid elongation1,

    an Arabidopsis gene required for the synthesis of VLCFAs found inseed triacylglycerols), involved in the biosynthesis of saturated and

    monounsaturated fatty acids with C1822+ chain length,[61].

    Until very recently, it was believed that FAE-like activities were

    restricted to only being involved in the synthesis of saturated and

    monounsaturated VLCFAs for use in wax and storage lipid synthe-

    sis. However, there is now evidence that this FAE-like class is also

    involved in the synthesis of VLC-PUFAs a PUFA-FAE was function-

    ally characterised from the parasitic protozoa Perkinsus marinus

    [62]. Interestingly, this KCS was in a small gene cluster with two

    cytochrome b5-fusion desaturases, and when all three open reading

    frames (ORFs) were heterologously expressed in yeast, the synthe-

    sis of ARA and EPA was achieved. As noted above, the heterologous

    activity of any KCS, be it ELO-like or FAE1-like, is dependent on the

    presence of the core elongase components. Although FAE1-likeKCSs are structurallyquite different to ELO-like (500aa, 2 TMs ver-

    sus

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    5/12

    short chain acyl-CoA and an unit of malonyl-CoA, followed by suc-

    cessive rounds of reduction, dehydration, reduction, and condensa-

    tion, with the acyl chain growing by two-carbon units with each

    round. A dehydratase/isomerase from this PKS complex catalyze

    the trans- tocis-conversion of the double bonds to form EPA and

    DHA. The domains encoding these activities are arranged sequen-

    tially on long (2030 kb) open reading frames (ORFs) in bacteria

    such as Shewanella andVibrio, and marine protist, as Schizochytrium.

    Their expression in yeast and plants has been reported [77] provid-

    ing an alternative to the aerobic desaturase/elongase system for

    transgenic VLC-PUFA production in plants. Interestingly, this PKS-

    like pathway generatesEPA or DHA as free fattyacids, meaning that

    these products will most likely requireactivation to CoAto facilitate

    their incorporation into lipids [78]. It is also worthy of note that

    genomic andbiochemical analysis of lipid biosynthesis in Schizochy-

    trium indicated that, in addition to thePKS-like pathway, this organ-

    ism also possessed desaturase and elongase activities of the

    D6-pathway, but crucially lacked the D12-desaturase activity

    (analogous to higher eukaryotes) [79]. The retention of this defec-

    tive aerobic pathway was suggested by the authors to represent a

    scavenging mechanism by which fatty acids prematurely released

    fromthe PKS-like system might undergo further modifications [79].

    4. Metabolic engineering to produce VLC-PUFAs in higher plants

    Higher plants lack the capacity to synthesize LC-PUFAs, thougha

    few taxonomically unrelated phyla can synthesize D6-desaturated

    fatty acids (thefirststepon theD6-pathway) such as omega-6c-lin-olenic acid (GLA; 18:3D6,9,12) and omega-3 stearidonic acid (SDA;

    18:4D6,9,12,15) [80]. The possibility of using transgenic plants that

    have been engineered to synthesize and accumulate VLC-PUFAs in

    their storage seed oils has been thoroughly investigated over the

    last 15 years. The conversion of native plant fatty acids such as LA

    and ALA to VLC-PUFAs requires a minimum of three sequential

    non-native enzymatic reactions (e.g. two desaturations and acyl-

    CoA elongation) to generate C20 PUFAs such as ARA and EPA. The

    last few years have seen considerable progress in the identificationfrom diverse sources (algae, fungi, mosses, plants and mammals) of

    genes encoding theprimary VLC-PUFA biosynthetic activities, effec-

    tively completingthe first stage of attempts to reverse-engineer this

    trait into a heterologous host such as a transgenic plant. Proof-of-

    concept demonstration that the VLC-PUFA pathway could function

    in a transgenic system was first provided by expression in yeast,

    with initial data showing the low accumulation of ARA and EPA

    [56,57,81,82]and subsequent experiments (with additional genes)

    to generate DHA [59]. Such experiments indicated the feasibility

    of transgenic expression of the LC-PUFA biosynthetic pathway in

    plants. The possibility of producing VLC-PUFAs in transgenic plants

    also became clear fromthe earliest attempts to accumulateGLA and

    SDA in oil-seed crops by expression of an individual gene, the

    D6-desaturase. Although the first expression of a cyanobacterialD6-desaturase in transgenic tobacco plants [83] resulted in the

    accumulation of low levels of these fatty acids, considerable pro-

    gress has been made reaching high levels (up to 40%) in several

    transgenic plants [49,84,85]. In 2004, three different reports by Qi

    et al. [86], Abbadi et al. [87]and Kinney et al. [88]demonstrated

    VLC-PUFAbiosynthesis in transgenicplantsby reverseengineering,

    although eachutilized distinctstrategies towardthe efficient recon-

    stitution of the process. Apart from the important biotechnological

    breakthroughs, they also provide some new insights into the bio-

    chemical pathways under manipulation and provide useful new

    tools for the dissection of the underlying enzymatic reactions.

    A diagrammatic summary of the results obtained from the stud-

    ies discussed below is shown inFig. 2.

    The first study utilizing the alternative pathway was the expres-sion of theIsochrysisC18 D9-elongase[89]in leaves ofArabidopsis

    using the constitutive CaMV 35S promoter and resulted in the syn-

    thesis of significant levels of EDA and ETriA (15% of total FA) [90].

    To fully reconstitute the alternative VLC-PUFA biosynthesis path-

    way for ARA and EPA, transgenic Arabidopsis lines expressing the

    Isochrysis D9-elongase were sequential transformed with the Eu-

    glena D8-desaturase and the M. alpina D5-desaturase [91] under

    the control of the constitutive 35S promoter [86]. ARA and EPA

    products were accumulated to a combined level of 10% (3% EPA

    and 6.6% ARA) of total fatty acids in leaf tissues; these data repre-

    sented an important proof-of-concept demonstration [86,92].

    Detailed analyses of leaf lipids[90]have confirmed that both D9-

    C18-PUFAs were efficiently elongated, accumulating to very high

    levels in the acyl-CoA pool of transgenic plants[93]. This indicated

    the inefficient transfer of these non-native fatty acids from the

    acyl-CoA pool into extra-plastidial phospholipids for their subse-

    quent desaturation. In addition to accumulation of ARA and EPA,

    several other C20 PUFA were also detected and identified as scia-

    donic acid (20:3D5,11,14) and juniperonic acid (20:4D5,11,14,17)

    [86]. These two non-methylene-interrupted PUFA appear to have

    arisen from the promiscuous activity of the D5-desaturase on

    substrates that might be expected to undergo D8-desaturation,

    due to a competition between enzymes for the elongated product.

    The M. alpina D5-desaturase used in the reconstitution of the alter-

    native VLC-PUFA biosynthetic pathway was previously observed to

    utilize unexpected substrates when individually expressed in

    transgenic canola, resulting in the accumulation of the unusual

    D5-desaturated C18 FA, taxoleic and pinolenic acids [91]. It re-

    mains to be seen how well the alternative pathway performs when

    it is expressed in seeds, as opposed to vegetative tissue.

    Complementary studies were described by Abbadi et al. [87]on

    the expression of the conventional D6-desaturase pathway in lin-

    seed (Linum usitatissimum) and tobacco by coexpressing the D5-

    and D6-desaturases from the diatom Phaeodactylum tricornutum

    [82] together with the D6-elongase from the moss Physcomitrella

    patens under the control of seed-specific promoters and introduced

    as a single integration event. Transgenic lines accumulated rela-

    tively low levels, only 1.6% EPA and 2.7% ARA. However, whilstthese C20 LC-PUFA were low, very high levels (>25% of total fatty

    acids) ofD6-desaturated fatty acids (GLA and SDA) were observed.

    This indicated that whilst the first desaturation in the VLC-PUFA

    biosynthetic pathway was functioning efficiently, the elongation

    step was severely limited. There was a bottleneck, described as

    substrate dichotomy [22], as a result of poor acyl-exchange of

    GLA and SDA from the phospholipid species from where they were

    generated to their acyl-CoA derivatives. The authors suggested that

    the linseed acyl-CoA:lyso-phosphatidylcholine acyltransferase

    (LPCAT), the enzyme believed to be primarily responsible in medi-

    ating acyl-exchange between phosphatidylcholine and the acyl-

    CoA pool [94], discriminates against D6-desaturated acyl groups

    as substrates. Detailed biochemical and metabolic analysis con-

    firmed that this poor exchange resulted in the incorporation ofGLA away from the VLC-PUFA biosynthetic activities, instead being

    directly incorporated into TAG in an acyl-CoA-independent man-

    ner. This is most likely to result from the direct conversion of phos-

    phatidylcholine (PC)-containing GLA to TAG, presumptively via a

    strong action of a phospholipid: diacylglycerol acyltransferase

    (PDAT)-like activity[87,95]. In respect to the substrate dichotomy

    bottleneck observed in linseed, this was analogous to that observed

    for the D8-alternative pathway in Arabidopsis [86], with the build-

    up of the product of the first enzyme in the pathway. This

    presumptively occurred as a result of poor acyl-exchange between

    the two metabolic pools (phospholipids, acyl-CoA) through which

    VLC-PUFA biosynthesis progresses, and highlights the importance

    of acyl-exchange in both the forward (acyl-CoA? PC; required

    after the first reaction of the D8-alternative pathway) andreverse (PC? acyl-CoA; required after the first reaction of the

    112 M. Venegas-Calern et al. / Progress in Lipid Research 49 (2010) 108119

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    6/12

    D6-pathway) directions in heterologous VLC-PUFA biosynthesis.

    Given that such acyl-exchange is dependent on endogenous acyl-transferases activities accepting non-native substrates (i.e. the

    intermediates of VLC-PUFA pathway) it is also likely that different

    activities (e.g. lyso-phospholipid acyltransferases, PDAT etc.) havedifferent affinities for these novel fatty acids[96]. Moreover, since

    Fig. 2. Overview of oil composition in transgenic lines. The fatty acid compositions of published transgenic lines have been compared, with the levels of target products and

    biosynthetic intermediates shown. The different configurations used are indicated. For clarity, the endogenous fatty acids are not shown, since these vary on a species-by-

    species basis. The studies compared are: Qi et al. (2004) [86], Abbadi et al. (2004)[87], Kinney et al. (2004) [88], Wu et al. 2005[100], Robert et al. (2005)[102], Hoffmann

    et al. (2008)[104]. (A) Omega-3 VLC-PUFA (EPA, DHA) accumulation in transgenic plants. (B) Omega-6 VLC-PUFA (ARA) accumulation in transgenic plants.

    M. Venegas-Calern et al./ Progress in Lipid Research 49 (2010) 108119 113

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    7/12

    many of these acyl-exchange enzymes can work in both forward

    and reverse directions, the pool sizes of individual metabolites is

    also likely to prove critical in determining the predominant en-

    zyme activity.

    A third exemplification is described in the patent application by

    Kinney et al. [88] realized in soybean (Glycine max). These

    researchers used a similar approach to that of Abbadi et al. [87],

    expressing genes encoding components of the conventional D6-

    desaturase pathway, a D6-desaturase (either from the oomycete

    fungusSaprolegnia diclinaorM. alpina), a D6-elongase from M. alp-

    ina and finally a D5-desaturase fromM. alpine, in transgenic soy-

    bean seeds and somatic embryos. However, to maximize the

    accumulation of omega-3 VLC-PUFAs such as EPA and DHA, anAra-

    bidopsis FAD3 gene[97]and aS. diclina D17-desaturase[98]were

    also co-expressed to turn the omega-6 PUFA metabolites into their

    omega-3 counterparts. The expression of these five enzymes

    yielded of 19.6% EPA in the transgenic somatic embryos, while al-

    most no ARA intermediate was observed because of the presence of

    the highly efficient D17-desaturase used. Although the reasons for

    the high yields obtained in soybean as compared to linseed are not

    clear, the differences between the two studies may be a reflection

    on the differing endogenous lipid metabolism present in linseed,

    though it should be noted that similar attempts to produce EPA

    in transgenic soybeans have been much less successful, for un-

    known reasons [99]. The lower accumulation of GLA in soybean

    oil, compared with tobacco and linseed, suggests a higher efficient

    transfer of GLA to the acyl-CoA pool for subsequent elongation by

    the reverse reaction of soybean LPCAT. Unexpectedly, up to 4.7% of

    x3-docosapentaenoic acid (DPA; 22:5D7,10,13,16, 19), a DHA pre-cursor, was also detected in the high EPA lines as a result of the

    additional activity of the M. alpine D6 elongase toward the D5-

    fatty acid EPA. This activity was not previously reported when

    the elongase was expressed in yeast [56],demonstrating a differ-

    ence in substrate-specificities in plant and yeast. This promiscuity

    is worthy of further investigation, and maybe indicates problems

    in the correct assembly of the elongase (discussed below). As a re-

    sult of the accumulation of DPA, Kinney et al. [88]carried out an-other co-transformation series with six cDNAs to produce DHA in

    somatic embryos. Additional genes for the D4-pathway, a D4-

    desaturase from the fungusSchizochytrium aggregatum and a spe-

    cific D5-elongase from the alga Pavlovasp. were expressed in addi-

    tion to the activities required to generate EPA. However, only low

    levels of DHA (2.03.3% of total fatty acids) were obtained, most

    likely due to the generic problems of substrate dichotomy dis-

    cussed above, resulting in inefficient acyl-exchange between the

    D5-elongase and the D4-desaturase.

    Two additional studies have demonstrated the accumulation of

    DHAandEPA inoilseeds. Firstly,Wu et al.[100] describedexpression

    oftheD6-pathwayin Brassica junceausing a similarapproachto that

    used by Kinney et al.[88]but involving more transgenes. A D17-

    desaturase from Phytophtora infestans was introduced to convertomega-6 substrates to omega-3 counterparts, a D12-desaturase

    from Calendula officinalis [101] toincrease theflux through theentire

    transgenicpathway andfinallya gene encoding a LPCAT from Thrau-

    stochytrium sp. was also co-expressed to increase exchange of D6-

    unsaturated acyl groups from acyl-phospholipids to acyl-CoAs for

    elongation. As a result of the genes co-expression transgenic B. jun-

    cea plants accumulated upto 25%ARA or 15%EPA [100]. This highle-

    vel of EPAallowed the introduction of the additional genes required

    forDHA synthesis(D5-elongase, D4-desaturase) to attempt thecon-

    version of EPA to DHA, resulting in low but significant amounts of

    DHA (0.21.5% of fatty acids in the seed lipids). These data indicate

    thatB. junceais a highly efficient host for the synthesis of ARA and

    EPA to high levels (comparable to that observed in soybean) but

    capable only of low level synthesis of DHA, reflecting an apparentblockin the conversionof EPA tothe C22 PUFA, DHA.Thiswouldalso

    indicatethat whilst endogenous B. juncea acyltransferases can facil-

    itate the exchange of acyl-intermediates on the pathway to EPA, the

    longer, more unsaturated forms of the DHA pathway are only very

    poorly utilized. Thus, the successful accumulation of DHA may re-

    quire the co-expression of suitable acyl-exchange activities.

    A similar approach was carried out by Robert et al. [102]

    expressing a bifunctional D6/D5-desaturase from zebrafish (Danio

    rerio)[103]in conjunction with theD6-elongase PEA-1 from C. ele-

    gans [57]to generate EPA. To generate DHA, two additional activi-

    ties (D5-elongase and D4-desaturase) from the algae Pavlova

    salinawere co-expressed [69]. Based on the observations of Abbadi

    et al.[87]and subsequent studies, Robert et al. hypothesised that

    the use of the (putative) acyl-CoA-dependent desaturase from zeb-

    rafish might overcome substrate dichotomy bottlenecks prior to

    D4-desaturation. However, whilst this study did demonstrate a

    proof-of-concept accumulation of ARA, EPAand DHAin Arabidopsis

    seeds, the levels achieved were relativelylow: ARA and EPA (4.2%of

    total lipids) and 0.20.5% of DHA. These results could be explained

    by lowsubstrate levels of LA-CoAand ALA-CoA in theacyl-CoA pool,

    which then rate-limits the levels ofD6-desaturation products and

    all subsequent metabolites. Alternatively, the codon usage of the

    two animal genes (C. elegans PEA-1 D6-ELO, D. rerio desaturase)

    may have resulted in the inefficient translation of these enzyme

    activities. Finally, it remains to be demonstrated that the D. rerio

    desaturase is indeed abona fide acyl-CoA dependent activity.

    These additional studies on the heterologous expression of

    desaturase/elongase combinations in different host plant species

    demonstrated that minor differences in host plant biochemistry

    can be of vital importance on the successful synthesis of ARA and

    EPA. Endogenous acyltransferases activities from transgenic soy-

    bean and Brassica presumptively have a broader substrate-specific-

    ity than linseed or tobacco and can partially overcome thesubstrate

    dichotomy problem. A further attempt to avoid the acyl-exchange

    bottleneck in transgenicplants by using acyl-CoA-dependent desat-

    uraseshasbeen recently described [104]. These authors isolatedand

    characterized two cDNAs from the microalga Mantoniella squamata

    which encoded for D6- and D5-desaturases with predictedacyl-CoA-substrate dependence (as described previously for a D6-

    desaturase from Ostreococcus tauri [105]. These desaturases were

    co-expressed under the control of a seed-specific promoter with

    the D6-elongase PSE1 of the moss P. patens [106] in Arabidopsis.

    Transgenic plants accumulated low but representative amounts of

    EPA, and crucially lacked the accumulation of D6-desaturation

    products previously observed in earlier studies. Thus, Hoffmann

    et al. [104]confirmed the potential of using acyl-CoA-dependent

    activities to overcome the problems associated with substrate

    dichotomy. However, it is perhaps surprising that this optimal

    configuration of the VLC-PUFA pathway yields only low levels of

    the target fatty acids (

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    8/12

    carrier protein(ACP) domains of theDHAsynthase PKS. These seeds

    accumulated up to 0.8% DHA with an additional 1.7% DPA n-6. Fur-

    ther optimizationof thispathway in commercial oilseeds is ongoing.

    5. Crucial issues: optimization the levels of LC-PUFA in

    transgenic plants

    Although the effective biosynthesis of ARA, EPA and to some ex-

    tent DHA has been demonstrated using different approaches in

    transgenic plants, the resultant fatty acid compositions and levels

    are not equivalent to that found in fish oil. Moreover, in most cur-

    rent examples such transgenic plants also contain high levels of

    omega-6 and omega-3 metabolic intermediaries. Marine oils, rich

    in EPA and/or DHA, are almost completely devoid of omega-6 fatty

    acids such as GLA and DHGLA. The goal now is to generate a veg-

    etal oil substitute for fish oils optimizing the accumulation of

    VLC-PUFAs. Several approaches (discussed below) have been sug-

    gested as a result of the studies described in this article. In addi-

    tion, these data also provide new insights into our understanding

    of plant lipid biochemistry, in particular the channelling of FA into

    various different lipids. Outlined below are logical approaches

    which might be expected to enhance the accumulation of VLC-PU-

    FAs in transgenic plants.

    5.1. The identification of superior desaturases

    The first approach is to identify highly active acyl-CoA depen-

    dent desaturases from a lower eukaryote, as has already been de-

    scribed for the D6-desaturase fromO. tauri [105]or M. squamata

    [104]. With such enzyme activities, both the desaturation and elon-

    gation reactions utilize acyl-CoA substrates and avoid the require-

    ment for acyl-exchange with PC. As noted above, published

    examples of the use of an acyl-CoA dependent route have resulted

    in the significant reduction in the accumulation of biosynthetic

    intermediates (most notably omega-6 GLA, linked to PC) but the ac-

    tual levels of target VLC-PUFAssuch as EPAand DHAare disappoint-

    ingly low [102,104]. This may be due to a number of problems(substrate availability, use of non-optimized sequences) or reflect

    additional (undefined) metabolic bottlenecks. It remains to be dem-

    onstrated that an exclusively acyl-CoA dependent pathway delivers

    significant improvement to yields of EPA or DHA levels, thought it

    could also be argued that the unambiguous identification of an

    acyl-CoA dependent D5-desaturase from lower eukaryotes is cur-

    rently lacking. Very recently, the molecular identification of an

    acyl-CoA dependent D12-desaturase was reported from insects

    [108]and it will be of interest to see if the co-expression of this

    activity would enhance (through the generation of LA-CoA) the

    activity of the algal acyl-CoA D6-desaturases in transgenic plants.

    A second modification based around desaturation is to ensure

    the conversion of omega-6 fatty acids to their omega-3 equivalents

    is through the use ofx3-desaturases: such enzyme activities havebeen demonstrated to be pivotal in the production of elevated lev-

    els of EPA in Brassica juncea [100]. Such x3-desaturases ideallyhave a high preference for C20 substrates (such as ARA) and have

    been identified from a number of fungal species[17]. An alterna-

    tive iteration is to identify VLC-PUFA desaturases with strong pref-

    erences for omega-3 substrates such as have been identified from

    M. squamata, Primula and Echium [104,109,110]. In the case of

    the Primula D6-desaturase, expression of this activity in linseed

    has recently been shown to result in the significant accumulation

    of SDA without the concomitant accumulation of GLA[85]. Another

    potentially very useful enzyme activity, a bifunctional D12- and

    D15-desaturase, has also recently been described from a number

    of organisms by several different groups. Damude et al.[111]iden-

    tified such a bifunctional desaturase fromFusarium moniliformeand demonstrated that the co-expression of this activity with the

    primary VLC-PUFA biosynthetic enzymes resulted in significant

    enhancement of the levels of EPA in both yeast (Y. lipolytica) and

    plants (soybean). Similarly, bifunctional desaturases were charac-

    terised from the free living amoeba A. castellanii [70], Claviceps

    purpurea [112] andCoprinus cinereus [113]. Thus, such activities

    have the potential to generate considerable omega-3 substrates

    for conversion to LC-PUFAs.

    5.2. Identification of a VLC-PUFA-specific acyl-exchange mechanism

    Metabolicbottlenecks appear to limit the full potential of oil-

    seed crops to accumulate economically sufficient amounts of these

    novel fatty acids. Such factors are likely to be represented by en-

    zymes involved in thechannelling andpartitioning of fatty acids be-

    tween the different metabolic pools involved in lipid synthesis and

    compartmentation this can take the form of spatial separation of

    different organelles (as is the case for TAG-containing oil bodies)

    or exchange between different substrate pools. Central to that latter

    problemis theso-called substrate dichotomy, where desaturation

    uses acyl-substrateslinkedto phospholipidswhereas elongation re-

    quires acyl-CoA substrates. It is predicted that the enzyme LPCAT,

    responsible for catalysing bidirectional exchange between these

    two pools, could help alleviate this bottleneck (represented sche-

    matically in Fig. 3). Very recently, genes encoding LPCAT have been

    functionally characterised from yeast and animals (reviewed in

    [114]), though in all cases only the forward reaction of LPCAT was

    demonstrated (i.e. acyl-CoA-dependent acylation of lyso-PC) (e.g.

    [115,116]). Interestingly, two Arabidopsis genes which showed

    strong homology to animal LPCATs were shown to encode predom-

    inantly LPEAT activities (acyl-CoA-dependent acylation of lyso-PE)

    [117]. Thus, the identification of a plant or algal form of LPCAT re-

    mains to be demonstrated, as does the role of the reverse reaction

    (release of a fatty acid from the sn-2 positions of PC and activation

    to acyl-CoA) and its utility in transgenic synthesis of VLC-PUFAs.

    5.3. Maintenance of a continuous flux of substrates through the

    VLC-PUFA biosynthetic pathway without significant loss to TAG

    Technological modifications to endogenous lipid metabolism to

    overcome such problem are not obvious, not least of all since such

    acyl-channelling represents the sum of multiple different acyl-

    exchange activities. In addition, it is highly likely that each plant

    G3PATG3P LPA PA DAG

    LPAAT PAPPC DAG

    DGATTAG

    PC pool

    CPT CPT

    Lyso-PC

    LPCAT

    Acyl-CoApool

    PDAT

    LPCAT

    Fig. 3. Schematic representation of triacylglycerol synthesis in plants. The acyl-

    CoA-dependent (Kennedy) pathway is shown as the central route for TAG synthesis.

    The primary activities are shown: acyl-CoA:glycerol-3-phosphate acyltransferases

    (G3PAT); acyl-CoA: lyso-phosphatidic acid acyltransferases (LPAAT); phosphatidic

    acid phosphatise (PAP); acyl-CoA:diacylglycerol acyltranserase (DGAT). Also shown

    are the acyl-CoA-independent activities such as the acyl-transfer between PC and

    DAG to generate TAG (catalysed by PDAT this reaction also generates lyso-PC) and

    also acyl-exchange between PC and DAG catalysed by cholinephosphotransferase

    (CPT). The acyl-CoA pool is generated by export of fatty acids from the plastid, and

    represents the sum of this activity plus reverse acyl-exchange from extra-plastidialphospholipids.

    M. Venegas-Calern et al./ Progress in Lipid Research 49 (2010) 108119 115

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    9/12

    species has a different combination of such activities (perhaps evi-

    denced by the huge disparity in the composition of plant seed oils),

    making a generic intervention unlikely if not impossible. A hypo-

    thetical solution might be the identification of TAGbiosynthetic en-

    zymes (such as diacylglycerol acyltransferases, DGAT or LPAT)

    which have a very strong substrate preferencefor EPAor DHA. How-

    ever, such activities are generally acyl-CoA-dependent, meaning

    that the substrate VLC-PUFA must be present in the acyl-CoA pool.

    The converse solution to this requirement is to use acyl-CoA-inde-

    pendent enzymes such PDAT[95], which catalyses the generation

    of TAG through the removal of fatty acids from the sn-2 position

    of phospholipids and then acylates them to DAG. Such an activity

    hastheadvantage of removing desaturation products fromtheirsite

    of synthesis, but it appears that PDATs are not restricted to PC as

    substrates. Genes for PDAT have been identified in plants, but little

    evidence has been found to date to suggest that these enzymes play

    a major quantitative or qualitative role in seed TAG metabolism.

    However, it may be possible to identify TAG biosynthetic enzymes

    from VLC-PUFA-synthesising lower eukaryotes which display the

    desired activities. Interestingly, recent evidence from plants has

    shown that the DGAT2 class of DGAT displays a more precise range

    of substrate-specificities, compared with DGAT1-type enzymes

    [118].

    5.4. Optimizing the fatty acid elongase

    Microsomal fatty acid elongation occurs as a result of four

    sequential enzymatic reactions: condensation, ketoreduction,

    dehydration andenoyl reduction, although for transgenic is possible

    through heterologous expression of just the initial condensing en-

    zyme [55,64]. It has been assumedthat the contribution of the other

    elongase components to VLC-PUFA synthesis is neutral, since the

    condensing enzyme acts in a trans-dominant manner. However, it

    is conceivable(if not likely) that the physical andbiochemical inter-

    actions between a non-native condensing enzyme and the other

    three endogenous elongase components may be sub-optimal. Per-

    haps of significance is the fact that in higher plants, unlike yeastor animals, the predominant microsomal KCS activities take the

    formof FAE1-like enzymes, whereas the transgenic VLC-PUFA elon-

    gating activity is of the ELO-like form [63]. In that respect the atyp-

    ical FAE1-like alternative pathway D9-elongating activity isolated

    fromP. marinus [62]may warrant further evaluation, as might the

    search for additional examples of FAE1-like VLC-PUFA elongating

    activities. Alternatively, the use of (ELO-like) activities from VLC-

    PUFA-synthesising lower plants such as Marchantia polymorpha

    may prove of benefit initial studies indicate that M. polymorpha

    activities perform well in transgenic plants [119]. Currently, our

    understanding of the regulation, organisation and assembly of the

    elongase complexis limited. Overexpression of theelongase ketore-

    ductaseresulted in an increasein theaccumulationof VLCmonosat-

    urated fatty acids in yeast, presumably by either increasing fluxthrough theelongase or increasingthe absolutenumber of elongase

    complexes, challenging the concept that the core elongase com-

    ponents have a neutral role in determining the levels of VLCFAs

    [120]. Thus, for all of the reasons outlined above, it may be that to

    obtain maximal elongation of target fatty acids, the additional core

    components of the elongases may need to be isolated from suitable

    EPA- or DHA-accumulating organisms and co-expressed with the

    transgene condensing enzyme from the same species.

    5.5. Modulating the acyl-CoA pool

    As discussed above, the use of acyl-CoA dependent desaturases

    is predicted to bypass the metabolic bottleneck generated by sub-

    strate dichotomy between the desaturase and the elongase. Thesuccess of this approach is dependent on significant levels of sub-

    strate fatty acids (LA, ALA) being present in the extra-plastidial

    acyl-CoA pool; given that the acyl-CoA pool in most plant cells is

    considered to be lower than that found in yeast or animals [121],

    this also indicates the requirement for a strong flux of fatty acids

    into this metabolic pool. One proven method for alteringthe profile

    of fatty acids present in the acyl-CoA pool is via the use of plastidial

    thioesterases which prematurely release fatty acids from the fatty

    acid synthase such approaches have been shown to generate in-

    creased levels of medium chain acyl-CoAs on expression of a Cuphea

    thioesterase [121]. However, it is not obvious how such approaches

    would directly result in the enhanced synthesis of VLC-PUFAs.

    Alternatively, it has recently been shown that blocking the peroxi-

    somal ABC transporter CTS (required for beta-oxidation) results in

    elevated levels of cytosolic acyl-CoAs and their incorporation into

    storage lipid [122]. Whilst a total blockade of beta-oxidation results

    in abnormal plant development and impaired germination, it may

    be possible to use developmentally-regulated silencing of such

    activities to modulate the acyl-CoA pool, increasing both the sub-

    strates available for VLC-PUFA biosynthesis and also the accumula-

    tion of target fatty acids in storage triacylglycerols. It must also be

    noted that our understanding of the flux of fatty acids into the acyl-

    CoA is partial, with very recent studies questioning the established

    model in which the products of the plastidial fatty acid synthase

    (16:0, 18:0, 18:1) are directly exported from the plastid into the

    cytosolic acyl-CoA pool [123]. Similarly, detailed kinetic analysis

    of the channelling of fatty acids into soybean embryo triacylglyce-

    rols indicates the central of acyl-exchange between PC and the

    acyl-CoA pool [124]. Thus, further research on the biosynthesis

    and homeostasis of the acyl-CoA pool is required.

    5.6. Co-ordinated expression of transgenes

    All of theexamples described above of theproduction of VLC-PU-

    FAs in transgenic plants have relied on the simultaneous co-expres-

    sion of desaturases and elongases in developing seeds. In most

    cases, the promoters used to drive this seed-specific expression

    were derived not from genes involved in oil biosynthesis but more

    often instead from storage protein synthesis. Thus, it may be possi-

    ble to enhance the overall levels of target VLC-PUFAs through the

    use of promoterswhose activity coincides with maximal oil synthe-

    sis and accumulation. Certainly the choice of appropriate promoter

    has been postulated to play a key role in the wide variation in VLC-

    PUFAs levels observed in transgenic soybeans[17,99].

    5.7. Appropriate localisation of transgene-derived activities

    It is now believed likely that many microsomal biochemical

    reactions occur in discrete sub-domains of the endomembrane sys-

    tem. Such sub-domains could be generated by local variation in li-

    pid compositions (such as so-called lipid rafts [125]) or via

    proteinprotein interaction to nucleate higher-order structures

    [126]. In either scenario, it is envisaged that multiple enzyme

    activities for a particular biochemical pathway are co-located,

    resulting in the minimal loss of intermediates and the optimal

    channelling of products to their intended metabolic pool. One pos-

    sible reason for limited production of non-native fatty acids such

    as VLC-PUFAs could be due to lack of sub-domain co-location for

    critical activities this could be either primary biosynthetic en-

    zymes or those involved in the generation of a strong flux (i.e. acyl-

    transferases such as DGAT[126].

    6. Conclusions and future perspectives

    It is obvious from the studies described in this article that het-erologous reconstitution of VLC-PUFA synthesis in transgenic

    116 M. Venegas-Calern et al. / Progress in Lipid Research 49 (2010) 108119

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    10/12

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    11/12

    [20] Napier JA, Beaudoin F, Michaelson LV, Sayanova O. The production of long

    chain polyunsaturated fatty acids in transgenic plants by reverse-

    engineering. Biochimie 2004;86:78592.

    [21] Sayanova OV, Napier JA. Eicosapentaenoic acid: biosynthetic routes and the

    potential for synthesis in transgenic plants. Phytochemistry 2004;65:14758.

    [22] Napier JA. The production of unusual fatty acids in transgenic plants. Ann Rev

    Plant Biol 2007;58:295319.

    [23] Nakamura MT, Nara TY. Structure, junction and dietary regulation of delta6,

    delta5 and delta9 desaturases. Ann Rev Nutr 2004;24:34576.

    [24] Nakamura MT, Nara TY. Essential fatty acid synthesis and its regulation in

    mammals. Prostagland Leukot Essent Fatty Acids 2003;68:14550.[25] Huang YS, Pereira SL, Leonard AE. Enzymes for transgenic biosynthesis of

    long-chain polyunsaturated fatty acids. Biochimie 2004;86:7938.

    [26] Cordain L, Eaton SB, Sebastian A, Mann N, Lindeberg S, Watkins BA, et al.

    Origins and evolution of the Western diet: health implications for the 21st

    century. Am J Clin Nutr 2005;81:34154.

    [27] Simopoulos AP. Essential fatty acids in health and chronic disease. Am J Clin

    Nutr 1999;70:560S9S.

    [28] Harris WS, Kris-Etherton PM, Harris KA. Intakes of long-chain omega-3 fatty

    acid associated with reduced risk for death from coronary heart disease in

    healthy adults. Curr Atheroscler Rep 2008;10:5039.

    [29] Horrobin DF. Interactions between n-3 and n-6 essential fatty acids (EFAs) in

    the regulation of cardiovascular disorders and inflammation. Prostagland

    Leukot Essent Fatty Acids 1991;44:12731.

    [30] Simopoulos AP. The importance of the ratio of omega-6/omega-3 essential

    fatty acids. Biomed Pharmacother 2002;56:36579.

    [31] Nugent AP. The metabolic syndrome. Nutr Bull 2004;29:3643.

    [32] Kris-Ethertona PM, Griegera JA, Ethertonb TD. Dietary reference intakes for

    DHA, EPA. Prostagland Leuk Essent Fatty Acids 2009;81:99104.

    [33] Calder PC, Yaqoob P. Omega-3 polyunsaturated fatty acids and human health

    outcomes. Biofactors 2009;35:26672.

    [34] Watts JL, Browse J. Genetic dissection of polyunsaturated fatty acid synthesis

    inCaenorhabditis elegans. Proc Natl Acad Sci USA 2002;99:58549.[35] Watts JL. Fat synthesis and adiposity regulation in Caenorhabditis elegans.

    Trends Endocrinol Metab 2009;20:5865.

    [36] Funk CD. Prostaglandins and leukotrienes: advances in eicosanoid biology.

    Science 2001;294:18715.

    [37] Tapiero H, Ba GN, Couvreur P, Tew KD. Polyunsaturated fatty acids (PUFA)

    and eicosanoids in human health and pathologies. Biomed Pharmacoth

    2002;56:21522.

    [38] Horrocks LA, Yeo YK. Health benefits of docosahexaenoic acid (DHA).

    Pharmacol Res 1999;40:21125.

    [39] Innis SM. The role of dietary n-6 and n-3 fatty acids in the developing brain.

    Dev Neurosci 2000;22:47480.

    [40] Voigt RG, Jensen CL, Fraley JK, Rozelle JC, Brown FR, Heird WC. Relationship

    between omega-3 long-chain polyunsaturated fatty acid status during early

    infancy and neurodevelopmental status at 1 year of age. J Hum Nutr Diet

    2000;15:11120.[41] Das UN. The lipids that matter from infant nutrition to insulin resistance.

    Prostagland Leuk Essent Fatty Acids 2002;67:112.

    [42] Rose DP, Connolly JM. Omega-3fatty acids as cancer chemopreventive agents.

    Pharmacol Ther 1999;83:21724.

    [43] Bell JG, Sargent JR, Tocher DR, Dick JR. Red blood cell fatty acid compositions

    in a patient with autistic spectrum disorder: a characteristic 32 abnormality

    in neurodevelopmental disorders? Prostagland Leuk Essent Fatty Acids

    2000;63:215.

    [44] Richardson AJ, Puri BK. A randomized double-blind, placebo-controlled study

    of the effects of supplementation with highly unsaturated fatty acids on

    ADHD-related symptoms in children with specific learning difficulties. Prog

    Neuro-Psychopharmacol Biol Psychiatry 2002;26:2339.

    [45] Morris MC, Evans DA, Bienias JL, Tangney CC, Bennett DA, Wilson RS, et al.

    Consumption of fish and n-3 fatty acids and risk incident Alzheimer disease.

    Arch Neurol 2003;60:9406.

    [46] Navarro E, Esteve M, Oliv A, Klaassen J, Cabr E, Tena X, et al. Abnormal fatty

    acid pattern in rheumatoid arthritis. A rationale for treatment with marine

    and botanical lipids. J Rheumatol 2000;27:298303.

    [47] Horrobin DF. Medical roles of metabolites of precursor EFAs. INFORM1995;6:42835.

    [48] Schwab JM, Chiang N, Arita M, Serhan CN. Resolvin E1 and protectin D1

    activate inflammation-resolution programmes. Nature 2007;447:86974.

    [49] Sayanova O, Smith MA, Lapinskas P, Stobart AK, Dobson G, Christie WW, et al.

    Expression of a borage desaturase cDNA containing an N-terminal

    cytochrome b5 domain results in the accumulation of high levels of 6-

    desaturated fatty acids in transgenic tobacco. Proc Natl Acad Sci USA

    1997;94:42116.

    [50] Napier JA, Sayanova O, Stobart AK, Shewry PR. A new class of cytochrome b5

    fusion proteins. Biochem J 1997;328:7178.

    [51] Stymne S, Appelqvist LA. The biosynthesis of linoleate from oleoyl-CoA via

    oleoyl-phosphatidylcholine in microsomes of developing safflower seeds. Eur

    J Biochem 1978;90:2239.

    [52] Griffiths G, Stobart AK, Stymne S. 6- and 12-desaturase activities and

    phosphatidic acid formation in microsomal preparations from the

    developing cotyledons of common borage (Borago officinalis). Biochem J1988;252:6417.

    [53] Jackson FM, Fraser TC, Smith MA, Lazarus C, Stobart AK, Griffiths G.Biosynthesis of C18 polyunsaturated fatty acids in microsomal membrane

    preparations from the filamentous fungusMucor circinelloides. Eur J Biochem1998;252:5139.

    [54] Fehling E, Lessire R, Cassagne C, Mukherjee KD. Solubilization and partial

    purification of constituents of acyl-CoA elongase from Lunaria annua. BiochimBiophys Acta 1992;1126:8894.

    [55] Millar AA, Kunst L. Very-long-chain fatty acid biosynthesis is controlled

    through the expression and specificity of the condensing enzyme. Plant J

    1997;12:12131.

    [56] Parker-Barnes JM, Das T, Bobik E, Leonard AE, Thurmond JM, Chuang L, et al.

    Identification and characterization of an enzyme involved in the elongation of

    n-6 and n-3 polyunsaturated fatty acid. Proc Natl Acad Sci USA2000;97:82849.

    [57] Beaudoin F, Michaelson L, Hey S, Lewis M, Shewry P, Sayanova O, et al.

    Heterologous reconstitution in yeast of the polyunsaturated fatty acid

    biosynthetic pathway. Proc Natl Acad Sci USA 2000;97:64216.

    [58] Oh CS, Toke DA, Mandala S, Martin CE. ELO2 and ELO3, homologues of the

    Saccharomyces cerevisiaeELO1 gene, function in fatty acid elongation and arerequired for sphingolipid formation. J Biol Chem 1997;272:1737684.

    [59] Meyer A, KirschH, Domergue F, Abbadi A, Sperling P, Bauer J, et al. Novelfatty

    acid elongases and their use for the reconstitution of docosahexaenoic acid

    biosynthesis. J Lipid Res 2004;45:1899909.

    [60] Qi B, Beaudoin F, Fraser T, Stobart AK, Napier JA, Lazarus CM. Identification of

    a cDNA encoding a novel C189 polyunsaturated fatty acid-specific

    elongating activity from the docosahexaenoic acid (DHA)-producing

    microalga,Isocarysis galbana. FEBS Lett 2002;510:15965.[61] James DW, Lim Jr E, Keller J, Plooy I, Ralston E, Dooner HK. Directed tagging of

    the Arabidopsis FATTY ACID ELONGATION1 (FAE1) gene with the maize

    transposon activator. Plant Cell 1995;7:30919.

    [62] Venegas-Calern M, Beaudoin F, Sayanova O, Napier JA. Cotranscribed genes

    for long chain polyunsaturated fatty acid biosynthesis in the protozoon

    Perkinsus marinus contain a plant-like FAE1 3-ketoacyl-CoA synthase. J BiolChem 2007;282:29963000.

    [63] Joubs J, Raffaele S, Bourdenx B, Garcia C, Laroche-Traineau J, Moreau P, et al.

    The VLCFA elongase gene family in Arabidopsis thaliana: phylogeneticanalysis, 3D modelling and expression profiling. Plant Mol Biol

    2008;67:54766.

    [64] Paul S, Gable K, Beaudoin F, Cahoon E, Jaworski J, Napier JA, et al. Members of

    the Arabidopsis FAE1-like 3-ketoacyl-CoA synthase gene family substitute for

    theElop proteins ofSaccharomyces cerevisiae. J Biol Chem 2006;281:901829.[65] Jakobsson A, Westerberg R, Jacobsson A. Fatty acid elongases in mammals:

    their regulation and roles in metabolism. Prog Lipid Res 2006;45:23749.

    [66] Wallis JG, Watts JL, Browse J. Polyunsaturated fatty acid synthesis: what will

    they think of next? Trends Biochem Sci 2002;27:467.

    [67] Korn ED. The fatty acids ofEuglena gracilis. J Lipid Res 1964;5:35262.[68] Wallis JG,Browse J. The 8-desaturaseofEuglena gracilis: an alternatepathway

    for synthesis of 20-carbon polyunsaturated fattyacids. Arch Biochem Biophys

    1999;365:30716.

    [69] Zhou XR, Robert SS, Petrie JR, Frampton DM, Mansour MP, Blackburn SI, et al.Isolation and characterization of genes from the marine microalgaPavlovasalina encodingthree front-end desaturases involved in docosahexaenoic acidbiosynthesis. Phytochemistry 2007;68:78596.

    [70] Sayanova O, Haslam R, Guschina I, Lloyd D, Christie WW, Harwood JL, et al. A

    bifunctional 12, 15-desaturase from Acanthamoeba castellanii directs thesynthesis of highly unusual n-1 series unsaturated fatty acids. J Biol Chem

    2006;281:3653341.

    [71] Qiu X. Biosynthesis of docosahexaenoic acid (DHA, 22:64, 7, 10, 13, 16, 19):

    twodistinct pathways. Prostagland Leukot Essent Fatty Acids 2003;68:1816.

    [72] Sprecher H, Chen Q, Yin FQ. Regulation of the biosynthesis of 22:5 n-6 and

    22:6 n-3: a complex intracellular process. Lipids 1999;34:S1536.

    [73] Meyer A, Cirpus P, Ott C, Schlecker R, Zahringer U, Heinz E. Biosynthesis of

    docosahexaenoic acid in Euglena gracilis: Biochemical and molecular evidencefor the involvement of a D4-fatty acyl group desaturase. Biochemistry

    2003;42:977988.

    [74] Pereira SL, Leonard AE, Huang YS, Chuang LT, Mukerji P. Identification of two

    novel microalgal enzymes involved in the conversion of the omega-3-fatty

    acid, eicosapentaenoic acid, into docosahexaenoic acid. Biochem J

    2004;384:35766.[75] Napier JA. Plumbing the depths of PUFA biosynthesis: a novel polyketide

    synthase-like pathway frommarine organisms. Trends Plant Sci 2002;7:514.

    [76] Metz JG, Roessler P, Facciotti D, Levering C, Dittrich F, Lassner M, et al.

    Production of polyunsaturated fatty acids by polyketide synthases in both

    prokaryotes and eukaryotes. Science 2001;293:2903.

    [77] Metz JG, Flatt JH, Kuner JM. PUFA polyketide synthase systems and uses

    thereof. Martek Biosciences Corporation WO 2006/135866 A2; 2006.

    [78] Metz JG,Kuner J, Rosenzweig B, Lippmeier JC, Roessler P, ZirkleR. Biochemical

    characterization of polyunsaturated fatty acid synthesis in Schizochytrium:release of the products as free fatty acids. Plant Physiol Biochem

    2009;47:4728.

    [79] Lippmeier JC, Crawford KS, Owen CB, Rivas AA, Metz JG, Apt KE.

    Characterization of both polyunsaturated fatty acid biosynthetic pathways

    inSchizochytrium sp.. Lipids 2009;44:62130.[80] Sayanova O, Napier JA, Shewry PR. 6-Unsaturated fatty acids in species and

    tissues of the Primulaceae. Phytochemistry 1999;52:41922.

    [81] Das T, Thurmond JM, Bobik E, Leonard AE, Parker-Barnes JM, Huang YS, et al.

    Polyunsaturated fatty acid-specific elongation enzymes. Biochem Soc Trans2000;28:65860.

    118 M. Venegas-Calern et al. / Progress in Lipid Research 49 (2010) 108119

  • 8/12/2019 An Alternative to Fish Oils Metabolic Engineering of Oil-seed Crops to Produce

    12/12

    [82] Domergue F, Lerchl J, Zahringer U, Heinz E. Cloning and functional char-

    acterization ofPhaeodactylum tricornutumfront-end desaturases involved ineicosapentaenoic acid biosynthesis. Eur J Biochem 2002;269:410513.

    [83] Reddy AS, Thomas TL. Expression of a cyanobacterial 6-desaturase gene

    results in gamma-linolenic acid production in transgenic plants. Nat

    Biotechnol 1996;14:63942.

    [84] Hong H, Datla N, Reed DW, Covello PS, MacKenzie SL, Qiu X. High-level

    production ofc-linolenic acid inBrassica juncea using a D6 desaturase fromPythium irregulare. Plant Physiol 2002;129:35462.

    [85] Ruiz-Lpez N, Haslam RP, Venegas-Calern M, Larson TR, Graham IA, Napier

    JA, et al. The synthesis and accumulation of stearidonic acid in transgenicplants: a novel source of heart-healthy omega-3fatty acids. Plant Biotechnol

    J 2009;7:70416.

    [86] Qi B, Fraser T, Mugford S, Dobson G, Sayanova O, Butler J, et al. Production of

    very long chain polyunsaturated omega-3 and omega-6 fatty acids in plants.

    Nat Biotechnol 2004;22:73945.

    [87] Abbadi A, Domergue F, Bauer J, Napier JA, Welti R, Zhringer U, et al.

    Biosynthesis of very-long-chain polyunsaturated fatty acids in transgenic

    oilseeds: constraints on their accumulation. Plant Cell 2004;16:273448.

    [88] Kinney AJ, Cahoon EB, Damude HG, Hitz WD, Kolar CW, Liu ZB. Production of

    very long chain polyunsaturated fatty acids in oilseed plants. WO 2004/

    071467 A2; 2004.

    [89] Qi B, Beaudoin F, Fraser T, Stobart AK, Napier JA, Lazarus CM. Identification of

    a cDNA encoding a novel C18-D9 polyunsaturated fatty acid-specific

    elongating activity from the docosahexaenoic acid (DHA)-producing

    microalga,Isochrysis galbana. FEBS Lett 2002;510:15965.[90] Fraser TCM, Qi B, Elhussein S, Chatrattanakunchai S, Stobart AK, Lazarus CM.

    Expression of the isochrysis C18-D9 polyunsaturated fatty acid specific

    elongase component alters arabidopsis glycerolipid profiles. Plant Physiol

    2004;135:85966.

    [91] Knutzon DS, Thurmond JM, Huang Y, Chaudhary S, Bobik Jr EG, Chan GM,

    et al. Identification of 5-desaturase from Mortierella alpina by heterologousexpression in bakers yeast and canola. J Biol Chem 1998;273:293606.

    [92] Green AG. From alpha to omega-producing essential fatty acids in plants. Nat

    Biotechnol 2004;22:6802.

    [93] Sayanova O, Haslam R, Qi B, Lazarus CM, Napier JA. The alternative pathway

    C20 8-desaturase from the non-photosynthetic organism Acanthamoebacastellanii is an atypical cytochrome b5-fusion desaturase. FEBS Lett2006;580:194652.

    [94] Stymne S, Stobart AK. Evidence for the reversibility of the acyl-

    CoA:lysophosphatidylcholine acyltransferase in microsomal preparations

    from developing safflower (Carthamus tinctorius L.) cotyledons and rat liver.Biochem J 1984;223:30514.

    [95] Dahlqvist A, Stahl U, Lenman M, Banas A, Lee M, Sandager L, et al.

    Phospholipid:diacylglycerol acyltransferase: an enzyme that catalyzes the

    acyl-CoA-independent formation of triacylglycerol in yeast and plants. Proc

    Natl Acad Sci USA 2000;97:648792.

    [96] Dyer JM, Stymne S, Green AG, Carlsson AS. High-value oils fromplants. Plant J2008;54:64055.

    [97] Yadav NS, Wierzbicki A, Aegerter M, Caster CS, Prez-Grau L, Kinney AJ, et al.

    Cloning of higher plant omega-3 fatty acid desaturases. Plant Physiol

    1993;103:46776.

    [98] Pereira SL, Huang YS, Bobik EG, Kinney AJ, Stecca KL, Packer JC, Mukerji P. A

    novel omega3-fatty acid desaturase involved in the biosynthesis of

    eicosapentaenoic acid. Biochem J 2004;378:66571.

    [99] Chen R, Matsui K, Ogawa M, Oe M, Ochiai M, Kawashima H. Expression ofD6,

    D5 desaturase and GLELO elongase genes from Mortierella alpina forproduction of arachidonic acid in soybean [Glycine max (L.) Merrill] seeds.Plant Sci 2006;170:399406.

    [100] Wu G, Truksa M, Datla N, Vrinten P, Bauer J, Zank T, et al. Stepwise

    engineering to produce high yields of very long-chain polyunsaturated fatty

    acids in plants. Nat Biotechnol 2005;23:10137.

    [101] Qiu X, Reed DW, Hong H, MacKenzie SL, Covello PS. Identification and

    analysis of a gene fromCalendula officinalis encoding a fatty acid conjugase.Plant Physiol 2001;125:84755.

    [102] Robert SS, Singh SP, Zhou X-R, Petrie JR, Blackburn SI, Mansour PM, et al.

    Metabolic engineering of Arabidopsis to produce nutritionally importantDHA in seed oil. Funct Plant Biol 2005;32:4739.

    [103] Hastings N, Agaba M, Tocher D, Leaver M, Dick J, Sargent JR, et al. A vertebrate

    fatty acid desaturase with D5 and D6 activities. Proc Natl Acad Sci USA

    2001;98:143049.

    [104] Hoffmann M, Wagner M, Abbadi A, Fulda M, Feussner I. Metabolic

    engineering of omega3-very long chain polyunsaturated fatty acid

    production by an exclusively acyl-CoA-dependent pathway. J Biol Chem

    2008;283:2235262.

    [105] Domergue F, Abbadi A, Zahringer U, Moreau H, Heinz E. In vivo

    characterization of the first acyl-CoA D6-desaturase from a member of the

    plant kingdom, the microalga Ostreococcus tauri. Biochem J2005;389:48390.

    [106] Zank TK, Zahringer U, Beckmann C, Pohnert G, Boland W, Holtorf H, et al.

    Cloning and functional characterisation of an enzyme involved in the

    elongation of 6-polyunsaturated fatty acids from the moss Physcomitrellapatens. Plant J 2002;31:25568.

    [107] Metz JG, Flatt JH, Kuner JM. The egenes for the enzymes of the

    polyunsaturated fatty acid polyketide synthase ofSchizochytrium and their

    use in the manufacture of polyunsaturated fatty acids. International PatentPublication WO 2006135866; 2006.

    [108] Zhou XR, Horne I, Damcevski K, Haritos V, Green A, Singh S. Isolation and

    functional characterization of two independently-evolved fatty acid 12-

    desaturase genes from insects. Insect Mol Biol 2008;17:66776.

    [109] Sayanova OV, BeaudoinF, Michaelson LV, Shewry PR, Napier JA. Identification

    of primula fatty acid 6-desaturases with n-3 substrate preferences. FEBS Lett

    2003;542:1004.

    [110] Garca-Maroto F, Maas-Fernndez A, Garrido-Crdenas JA, Alonso DL.

    Substrate specificity of acyl-6-desaturases from Continental versus

    Macaronesian echiumspecies. Phytochemistry 2006;67:5404.[111] Damude HG, Zhang H, Farrall L, Ripp KG, Tomb J-F, Hollerbach D, et al.

    Identification of bifunctional 12/3 fatty acid desaturases for improving the

    ratio of 36 fatty acids in microbes and plants. PNAS 2006;103:944651.

    [112] Meesapyodsuk D, Reed DW, Covello PS, Qiu X. Primary structure,

    regioselectivity, and evolution of the membrane-bound fatty acid

    desaturases ofClaviceps purpurea. J Biol Chem 2007;282:201919.[113] Zhang S, Sakuradani E, Ito K, Shimizu S. Identification of a novel bifunctional

    12/15 fatty acid desaturase from a basidiomycete,Coprinus cinereusTD#8222. FEBS Lett 2007;581:3159.

    [114] Sthl U, Stlberg K, Stymne S, Ronne H. A family of eukaryotic

    lysophospholipid acyltransferases with broad specificity. FEBS Lett

    2008;582:3059.

    [115] Chen X, Hyatt BA, Mucenski ML, Mason RJ, Shannon JM. Identification and

    characterization of a lysophosphatidylcholine acyltransferase in alveolar type

    II cells. Proc Natl Acad Sci USA 2006;103:117249.

    [116] Nakanishi H, Shindou H, Hishikawa D, Harayama T, Ogasawara R, Suwabe A,

    et al. Cloning and characterization of mouse lung-type acyl-

    CoA:lysophosphatidylcholine acyltransferase 1 (LPCAT1). Expression in

    alveolar type II cells and possible involvement in surfactant production. J

    Biol Chem 2006;281:201407.

    [117] Stlberg K, Sthl U, Stymne S, Ohlrogge J. Characterization of two Arabidopsis

    thaliana acyltransferases with preference for lysophosphatidylethanolamine.

    BMC Plant Biol 2009;9:60.

    [118] Burgal J, Shockey J, Lu C, Dyer J, Larson T, Graham I, et al. Metabolic

    engineering of hydroxy fatty acid production in plants: RcDGAT2 drives

    dramatic increases in ricinoleate levels in seed oil. Plant Biotechnol J

    2008;6:81931.[119] Kajikawa M, Matsui K, Ochiai M, Tanaka Y, Kita Y, Ishimoto M, et al.

    Production of arachidonic and eicosapentaenoic acids in plants using

    bryophyte fatty acid 6-desaturase, 6-elongase, and 5-desaturase genes.

    Biosci Biotechnol Biochem 2008;72:43544.

    [120] Beaudoin F, Gable K, Sayanova O, Dunn T, Napier JA. A Saccharomycescerevisiaegene required for heterologous fatty acid elongase activity encodesa microsomal beta-keto-reductase. J Biol Chem 2002;277:114818.

    [121] Larson TR, Graham IA. A novel technique for the sensitive quantification of

    acyl CoA esters from plant tissues. Plant J 2001;25:11525.

    [122] Slocombe SP, Cornah J, Pinfield-Wells H, Soady K, Zhang Q, Gilday A, et al. Oil

    accumulation in leaves directed by modification of fatty acid breakdown and

    lipid synthesis pathways. Plant Biotechnol J 2009;7:694703.

    [123] Bates PD, Ohlrogge JB, Pollard M. Incorporation of newly synthesized fatty

    acids into cytosolic glycerolipids in pea leaves occurs via acyl editing. J Biol

    Chem 2007;282:3120616.

    [124] Bates PD, Durrett TP, Ohlrogge JB, Pollard M. Analysis of acyl fluxes through

    multiple pathways of triacylglycerol synthesis in developing soybean

    embryos. Plant Physiol 2009;150:5572.

    [125] Borner GH, Sherrier DJ, Weimar T, Michaelson LV, Hawkins ND, Macaskill A,et al. Analysis of detergent-resistant membranes in Arabidopsis. Evidence for

    plasma membrane lipid rafts. Plant Physiol 2005;137:10416.

    [126] DyerJM, Mullen RT. Engineering plantoils as high-value industrialfeedstocks

    for biorefining: the need for underpinning cell biology research. Physiol Plant

    2008;132:1122.

    [127] Kalaitzandonakes N, Alston JM, Bradford KJ. Compliance costs for regulatory

    approval of new biotech crops. Nat Biotechnol 2007;25:50911.

    [128] Cressey D. Aquaculture: future fish. Nature 2009;458:398400.

    M. Venegas-Calern et al./ Progress in Lipid Research 49 (2010) 108119 119