WO2008025068A1 - Synthesis of fatty acids - Google Patents

Synthesis of fatty acids Download PDF

Info

Publication number
WO2008025068A1
WO2008025068A1 PCT/AU2007/001242 AU2007001242W WO2008025068A1 WO 2008025068 A1 WO2008025068 A1 WO 2008025068A1 AU 2007001242 W AU2007001242 W AU 2007001242W WO 2008025068 A1 WO2008025068 A1 WO 2008025068A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
seq
desaturase
cell
fatty acid
Prior art date
Application number
PCT/AU2007/001242
Other languages
French (fr)
Inventor
Katherine Damcevski
Karen Glover
Allan Green
Victoria S. Haritos
Irene Horne
Surinder Pal Singh
Craig C. Wood
Xue-Rong Zhou
Original Assignee
Commonwealth Scientific And Industrial Research Organisation
Grains Research And Development Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Commonwealth Scientific And Industrial Research Organisation, Grains Research And Development Corporation filed Critical Commonwealth Scientific And Industrial Research Organisation
Priority to AU2007291937A priority Critical patent/AU2007291937B2/en
Priority to CA002661697A priority patent/CA2661697A1/en
Priority to BRPI0716075-5A priority patent/BRPI0716075A2/en
Priority to US12/310,645 priority patent/US8816106B2/en
Priority to EP07784864A priority patent/EP2059588A4/en
Publication of WO2008025068A1 publication Critical patent/WO2008025068A1/en
Priority to US14/467,942 priority patent/US10513717B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/64Fats; Fatty oils; Ester-type waxes; Higher fatty acids, i.e. having at least seven carbon atoms in an unbroken chain bound to a carboxyl group; Oxidised oils or fats
    • C12P7/6409Fatty acids
    • C12P7/6427Polyunsaturated fatty acids [PUFA], i.e. having two or more double bonds in their backbone
    • CCHEMISTRY; METALLURGY
    • C11ANIMAL OR VEGETABLE OILS, FATS, FATTY SUBSTANCES OR WAXES; FATTY ACIDS THEREFROM; DETERGENTS; CANDLES
    • C11BPRODUCING, e.g. BY PRESSING RAW MATERIALS OR BY EXTRACTION FROM WASTE MATERIALS, REFINING OR PRESERVING FATS, FATTY SUBSTANCES, e.g. LANOLIN, FATTY OILS OR WAXES; ESSENTIAL OILS; PERFUMES
    • C11B1/00Production of fats or fatty oils from raw materials
    • C11B1/10Production of fats or fatty oils from raw materials by extracting
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8241Phenotypically and genetically modified plants via recombinant DNA technology
    • C12N15/8242Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits
    • C12N15/8243Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits involving biosynthetic or metabolic pathways, i.e. metabolic engineering, e.g. nicotine, caffeine
    • C12N15/8247Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits involving biosynthetic or metabolic pathways, i.e. metabolic engineering, e.g. nicotine, caffeine involving modified lipid metabolism, e.g. seed oil composition
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0071Oxidoreductases (1.) acting on paired donors with incorporation of molecular oxygen (1.14)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/64Fats; Fatty oils; Ester-type waxes; Higher fatty acids, i.e. having at least seven carbon atoms in an unbroken chain bound to a carboxyl group; Oxidised oils or fats
    • C12P7/6409Fatty acids
    • C12P7/6427Polyunsaturated fatty acids [PUFA], i.e. having two or more double bonds in their backbone
    • C12P7/6431Linoleic acids [18:2[n-6]]

Definitions

  • the present invention relates to enzymes which possess desaturase, conjugase, epoxidase, and/or hydroxylase activity that can be used in methods of synthesizing fatty acids.
  • the primary products of fatty acid biosynthesis in most organisms are 16- and 18-carbon compounds.
  • the relative ratio of chain lengths and degree of unsaturation of these fatty acids vary widely among species. Mammals and insects, for example, produce primarily saturated and monosaturated fatty acids, while most higher plants produce fatty acids with one, two, or three double bonds, the latter two comprising polyunsaturated fatty acids (PUF A's).
  • PEF A's polyunsaturated fatty acids
  • omega-3 fatty acids also represented as “n-3” fatty acids
  • EPA eicosapentaenoic acid
  • omega-6 fatty acids also represented as “n-6” fatty acids
  • ARA arachidonic acid
  • the ability of cells to modulate the degree of unsaturation in their membranes is mainly determined by the action of fatty acid desaturases.
  • Desaturase enzymes introduce unsaturated bonds at specific positions in their fatty acyl chain substrates. Desaturase enzymes generally show considerable selectivity both for the chain length of the substrate and for the location of existing double bonds in the fatty acyl chain (Shanklin and Cahoon, 1998) and may be classified on this basis.
  • Another classification of fatty acid desaturases is based on the moiety to which the hydrocarbon chains of their substrates are acylated. Desaturases recognize substrates that are bound either to acyl carrier protein, to coenzyme A, or to lipid molecules such as phospholipids (Murata and Wada, 1995; Shanklin and Cahoon, 1998).
  • the desaturation of fatty acids in glycerolipids is essential for the proper function of biological membranes.
  • Introduction of unsaturation in the ⁇ 9 position of palmitic or stearic acid provides fluidity to membrane lipids and thus ⁇ 9 desaturases are found universally in living systems.
  • Linoleic acid (LA; 18:2, ⁇ 9, 12) is produced from oleic acid (18:1, ⁇ 9) by a ⁇ 12-desarurase while ⁇ -linolenic acid (ALA; 18:3) is produced from LA acid by a ⁇ 15-desaturase.
  • Stearidonic acid (18:4, ⁇ 6, 9, 12, 15) and ⁇ -linolenic acid (18:3, ⁇ 6, 9, 12) are produced from ALA and LA, respectively, by a ⁇ 6-desaturase.
  • mammals cannot desaturate beyond the ⁇ 9 position and therefore cannot convert oleic acid into LA.
  • eukaryotes including fungi, nematodes and plants, have enzymes which desaturate at the carbon 12 and carbon 15 positions.
  • the membrane-associated ⁇ 12 desaturases of Arahidopsis sp. and soybean use acyl lipid substrates
  • saturated fatty acids can be unsaturated initially at positions other than the 9-position by desaturases with unusual specificities.
  • Petroselinic acid cis-6-octadecenoic acid
  • Umbelliferae Apiaceae
  • Araliaceae Araliaceae
  • Garryaceae plant families, where it can reach 85% of the total lipid fatty acid (Kleiman and Spencer, 1982).
  • the desaturase from coriander (Coriandrum sativum) has been characterised that makes the unusual lipid.
  • a plastid- located acyl-acyl carrier protein (ACP) ⁇ 4 desaturase acts on ACP -palmitic acid to produce cis-4-hexadecenoic acid which is transferred from the plastid to the developing seed where it is elongated to cis-6-octadecenoic acid (Cahoon et al. 1992).
  • a related desaturase with 83% sequence identity has been obtained from English Ivy (Hedra helix L.) which produces cis-4-hexadecenoic acid and cis-6-octadecenoic acid when expressed in Arabidopsis (Whittle et al 2005).
  • cis-5- Eicosenoic acid (C20:l ⁇ 5) is a major component of the seed oil of meadowfoam (Limnanthes alba) and related Limnathes species.
  • the enzyme responsible for the production of the unusual oil in L. douglasii was identified as an acyl coenzyme A- ⁇ 5 desaturase whose substrate preference is eicosanoic acid (Cahoon et al., 2000). Expression of L.
  • Genes encoding similar enzymes have not been cloned from animals such as insects.
  • Some Lepidopteran insects carry out ⁇ l l desaturation of saturated fatty acids (C16:O, C18:0) esterified to acylCoA as a step in the production of a diversity of moth sex pheromones (Rodriguez et al 2004).
  • Desaturases from the moth Spodoptera littoralis were expressed in Saccharomyces cerevisiae to produce cis- ⁇ l l mono- unsaturated products of C14:0, C16:0 and C18:0 when the yeast cells were fed additional saturated fatty acids (Rodriguez et al 2004).
  • trans- ⁇ l l tetradecenoic acid was formed from myristic acid (C 14:0) fed to the yeast.
  • a minor byproduct of the ⁇ l l desaturation was the formation of 11 -hydroxy hexadecanoic or octadecanoic acid (up to 0.1% of total fatty acids) (Serra et al., 2006).
  • Moto et al. (2004) identified a bi-functional acyl-CoA desaturase from the pheromone gland of the silkmoth which was responsible for the biosynthesis of the pheromone precursor.
  • the desaturase first utilised palmitic acid to make cis-11-hexadecenoic acid and then acted on this to remove allylic 2H and form a conjugated diene fatty acid (trans- ⁇ 10,cis- ⁇ 12-hexadecendienoic acid and some trans- ⁇ 10,trans- ⁇ 12-hexadecendienoic acid) and therefore it possessed both cis- ⁇ l l and conjugase desaturase activities, hi the New Zealand leaf roller, Pl ⁇ notortrix octo, a desaturase has been identified from pheromone gland that desaturates palmitic acid at the ⁇ 10 position to form cis- ⁇ lO- hexadecenoic acid (Hao et al. 2002).
  • Omega-3 LC-PUFA are now widely recognized as important compounds for human and animal health and the inclusion of omega-3 LC-PUFA such as EPA and DHA in the human diet has been linked with numerous health-related benefits. These include prevention or reduction of coronary heart disease, hypertension, type-2 diabetes, renal disease, rheumatoid arthritis, ulcerative colitis, chronic obstructive pulmonary disease, various mental disorders such as schizophrenia, attention deficit hyperactive disorder and Alzheimer's disease, and aiding brain development and growth (Simopoulos, 2000).
  • fatty acids may be obtained from dietary sources or by conversion of linoleic (LA, omega-6) or ⁇ -linolenic (ALA, omega-3) fatty acids, both of which are regarded as essential fatty acids in the human diet. While humans and many other vertebrate animals are able to convert LA or ALA, obtained from plant sources, to LC-PUFA, they carry out this conversion at a very low rate. Moreover, most modern societies have unbalanced diets in which at least 90% of polyunsaturated fatty acid(s) consist of omega-6 fatty acids, instead of the 4:1 ratio or less for omega-6: omega-3 fatty acids that is regarded as ideal (Trautwein, 2001).
  • LC-PUFA eicosapentaenoic acid
  • DHA 5 22:6 docosahexaenoic acid
  • Fatty acids may also be hydroxylated, for example ricinoleic acid (12-hydroxy- octadec-cw-9-enoic acid) which comprises up to 90% of the fatty acid in castor oil from Ricinus communis and is an important agricultural commodity oil.
  • ricinoleic acid (12-hydroxy- octadec-cw-9-enoic acid) which comprises up to 90% of the fatty acid in castor oil from Ricinus communis and is an important agricultural commodity oil.
  • Other related hydroylated fatty acids found in plant oils include 12-hydroxy-octadeca-c «-9,cz,s' ⁇ 15- dienoic (densipolic) and 14-hydroxy-eicosa-cw-ll,cw-17-dienoic (auricolic) acids.
  • the Ricinus ⁇ 12 hydroxylase acts on oleic acid lipid substrate to produce ricinoleic acid; the desaturase gene responsible for the transformation is most closely related to but divergent from plant membrane ⁇ 12 acyl lipid desaturases (van de Loo et al 1995).
  • the L.fendleri hydroxylase gene has been cloned and expressed in an Arabidopsis FAD2 mutant which accumulated ricinoleic, lesquerolic and densipolic acids in seeds (Broun et al 1998).
  • 2-hydroxy fatty acids occur in appreciable amounts in the sphingolipids of plants and animals but they are also present as minor components of seeds oils such as 2-hydroxy-octadeca-9,12,15- trienoate from Thymus vulgaris seeds and 2-hydroxy-oleic and linoleic acids are found in Salvia nilotica (Smith, 1971; Badami and Patil, 1981).
  • Fatty acids may also comprise epoxy groups.
  • the most widely known natural epoxy fatty acid is vernolic acid (12,13-epoxy-octadec-cw-9-enoic acid) from the seed oils of Vernonia spp and Euphorbia lagascae (Cuperus and Derksen, 1996).
  • the epoxygenase gene of C. palaestina which is related to but divergent from plant membrane ⁇ 12-oleate desaturases, has been functionally characterized and shown to use linoleate as a substrate (Lee et al. 1998).
  • conjugated fatty acids are produced by the activity of a conjugase (Crombie et al., 1984; Crombie et al., 1985; Fritsche et al., 1999; Cahoon et al., 2001; Qiu et al., 2001).
  • conjugated fatty acids such as calendulic acid, eleostearic acid or punicic acid proceeds via the desaturation of oleic acid to linoleic acid by a ⁇ 12-desaturase and a further desaturation in conjunction with a rearrangement of the Z9- or Z12-double bond to the conjutrienic fatty acid by a specific conjutriene-forming desaturase (conjugase).
  • the present inventors have identified novel enzymes having desaturase, conjugase, epoxygenase, and/or hydroxylase activity from insects of the Order Coleoptera and Orthoptera.
  • the present invention provides a eukaryotic cell comprising an exogenous nucleic acid encoding a polypeptide which is:
  • polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and/or 134, and/or
  • polypeptide (iii) a biologically active fragment of i) or ii), wherein the polypeptide has one or more activities selected from desaturase, conjugase, epoxidase and hydroxylase activity.
  • the polypeptide can be isolated from an insect of the Order Coleoptera or Orthoptera.
  • insect species from which the polypeptide can be isolated include, but are not limited to, Tribolium, Chauliognathus or Acheta.
  • polypeptide encoded by the exogenous nucleic acid is:
  • polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134, and/or
  • polypeptide is:
  • polypeptide comprising amino acids having a sequence which is at least 50% identical to a sequence set forth in SEQ ID NO: 18 or SEQ ID NO: 19, and/or
  • polypeptide is:
  • polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs: 17, 22, 23, 27, 29, 74, 75 and/or 76
  • polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 17, 22, 23, 27, 29, 74, 75 and/or 76, and/or
  • the present inventors are the first to identify a nucleic acid encoding an an acyl-CoA ⁇ 12 desaturase.
  • the present invention provides a eukaryotic cell comprising an exogenous nucleic acid encoding an acyl-CoA ⁇ 12 desaturase.
  • the acyl-CoA ⁇ 12 desaturase comprises:
  • amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134, and/or
  • the eukaryotic cell comprises an increased level of 16:2 ⁇ 9 ' ⁇ 12 and/or 18:2 ⁇ 9 ' A12 fatty acids relative to a corresponding eukaryotic cell lacking the exogenous nucleic acid.
  • the eukaryotic cell comprises an increased level of 16:2 ⁇ 9 ' ⁇ 12 and/orl8:2 ⁇ 9 ' ⁇ 12 fatty acids which are esterified to CoA.
  • the present inventors are also the first to identify an acyl-CoA ⁇ 5 desaturase which has a preference for a 18:0 and/or 16:0 fatty acid substrate when compared to a number of other substrates. Accordingly, in a further aspect the present invention provides a eukaryotic cell comprising an exogenous nucleic acid encoding an acyl- CoA ⁇ 5 desaturase, wherein the desaturase is more active on a 18:0 and/or 16:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is any one, two, three or all of a 18:1 ⁇ 9 , 16:1 A9 , 20:0 and 20:2 ⁇ 11 ⁇ 14 .
  • the acyl-CoA ⁇ 5 desaturase comprises:
  • the eukaryotic cell comprises an increased level of 16:1 5 and/or 18:1 A5 fatty acids relative to a corresponding eukaryotic cell lacking the exogenous nucleic acid.
  • the eukaryotic cell comprises an increased level of 16:1 ⁇ 5 and/orl8:l ⁇ 5 fatty acids which are esterified to CoA.
  • the present inventors are also the first to identify an acyl-CoA ⁇ 9 desaturase which is more active on a 14:0 substrate than on certain fatty acid substrates esterified to CoA.
  • the present invention provides a eukaryotic cell comprising an exogenous nucleic acid encoding an acyl-CoA ⁇ 9 desaturase, wherein the desaturase is more active on a 14:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is 16:0 and/or 18:0.
  • the acyl-CoA ⁇ 9 desaturase comprises:
  • the eukaryotic cell comprises an increased level of 14:1 A9 relative to a corresponding eukaryotic cell lacking the exogenous nucleic acid.
  • the eukaryotic cell comprises an increased level of 14:1 ⁇ 9 which is esterified to CoA.
  • the eukaryotic cell comprising the exogenous nucleic acid is a plant cell, a mammalian cell, an insect cell, a fungal cell or a yeast cell.
  • the eukaryotic cell is in a plant or plant seed.
  • the plant or plant seed is an oilseed plant or an oilseed respectively.
  • a process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
  • nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence more closely related to SEQ ID NO: 28 than to SEQ ID NO: 135,
  • the present invention provides a process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
  • nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence that is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134,
  • step (iv) comprises selecting a nucleic acid molecule encoding an acyl-CoA ⁇ 12 desaturase.
  • the modified fatty acid composition comprises an increased level of 16:2 ⁇ 9 ' ⁇ 12 and/or 18 :2 ⁇ 9 ⁇ 12 .
  • the present invention further provides a process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
  • nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence that is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 18 and/or 19,
  • step (iv) comprises selecting a nucleic acid molecule an acyl-CoA ⁇ 5 desaturase, wherein the desaturase is more active on a 18:0 and/or 16:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is any one, two, three or all of a 18:1 ⁇ 9 , 16:1 ⁇ 9 , 20:0 and 20:2 ⁇ 11 ⁇ 14 .
  • the modified fatty acid composition comprises an increased level of 16:1 ⁇ 5 and/or 18:1 ⁇ 5 fatty acids.
  • the present invention further provides a process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
  • nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence that is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 17, 22, 23, 27, 29, 74, 75 and/or 76, (ii) introducing the nucleic acid molecule into a cell or cell-free expression system in which the promoter is active,
  • step (iv) comprises selecting a nucleic acid molecule encoding an acyl-CoA ⁇ 9 desaturase, wherein the desaturase is more active on a 14:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is 16:0 and/or 18:0.
  • the modified fatty acid composition comprises an increased level of 14: 1 ⁇ 9 .
  • polypeptide encoded by the nucleic acid molecule is an insect polypeptide or mutant thereof.
  • the present invention further provides a substantially purified or recombinant polypeptide which is an acyl-CoA ⁇ 12 desaturase.
  • polypeptide is:
  • polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134, and/or
  • the present invention further provides a substantially purified or recombinant polypeptide which is an acyl-CoA ⁇ 5 desaturase, wherein the desaturase is more active on a 18:0 and/or 16:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is any one, two, three or all of a 18: 1 ⁇ 9 , 16:1 ⁇ 9 , 20:0 and
  • polypeptide is:
  • polypeptide comprising amino acids having a sequence which is at least 50% identical to SEQ ID NO: 18 and/or SEQ ID NO: 19, and/or
  • the present invention further provides a substantially purified or recombinant polypeptide which is an acyl-CoA ⁇ 9 desaturase, wherein the desaturase is more active on a 14:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is 16:0 and/or 18:0.
  • the polypeptide is:
  • polypeptide comprising amino acids having a sequence which is at least 50% identical to one or more of the sequences as set forth in SEQ ID NO: 17 and/or SEQ ID NO: 74, and/or
  • the present invention further provides a substantially purified or recombinant polypeptide which is:
  • polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and/or 134, and/or
  • polypeptide (iii) a biologically active fragment of i) or ii), wherein the polypeptide has one or more an activities selected from desaturase, conjugase, epoxidase and/or hydroxylase activity.
  • the polypeptide comprises amino acids having a sequence which is at least 90% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and/or 134.
  • polypeptide can be isolated from an insect of the Order Coleoptera or Orthoptera.
  • the polypeptide has desaturase activity upon a carbon-carbon bond located at any one of the ⁇ 2 to ⁇ 15 positions of a fatty acid.
  • the polypeptide has desaturase activity on a C16 or C18 fatty acid.
  • polypeptide is a fusion protein further comprising at least one other polypeptide sequence.
  • the present invention further provides an isolated and/or exogenous polynucleotide comprising:
  • a vector comprising a polynucleotide of the invention.
  • the polynucleotide is operably linked to a promoter.
  • the present invention further provides a cell comprising the recombinant polypeptide according to the invention, the exogenous polynucleotide of the invention and/or the vector of the invention.
  • the cell is a plant, fungal, yeast, bacterial or animal cell. More preferably, the cell is a eukaryote cell.
  • the present invention further provides a method of producing the polypeptide according to the invention, the method comprising expressing in a cell or cell free expression system the polynucleotide of the invention.
  • the method further comprises isolating the polypeptide.
  • the present invention further provides a transgenic non-human organism comprising a cell according to the invention.
  • the organism is a transgenic plant.
  • the present invention further provides a seed comprising the cell according to the invention.
  • the present invention further provides oil produced by, or obtained from, the cell according to the invention, the transgenic non-human organism of the invention, or the seed of the invention.
  • the oil comprises fatty acids 16:0, 16:1 A5 , 18:0 and 18:1 ⁇ 5 , wherein the ratio of the total amount of 18: 1 ⁇ 5 to 18:0 in the oil is between 100:1 and 1:2, and wherein the fatty acid of the oil comprises less than 10%, or less than 5% (w/w) 20:l ⁇ 5 .
  • At least 43%, or at leasst 50%, or at least 60%, of the C18 fatty acid of the oil is 18 :1 ⁇ 5 .
  • the fatty acid of the oil comprises at least 3.0% (w/w), or at least 5% (w/w) or at least 10% (w/w), 18 : 1 ⁇ 5 as a percentage of the total fatty acid of the oil.
  • the oil comprises less than 10% (w/w), or less than 5% (w/w), 18:3 ⁇ 9 ⁇ 12 ' ⁇ 15 (ALA) as a percentage of the total fatty acid in the oil.
  • the oil is obtained by extraction of oil from an oilseed.
  • the present invention further provides an oil comprising fatty acids 16:0, 16:1 ⁇ 5 , 18:0 and 18:1 ⁇ 5 , wherein the ratio of the total amount of 18:1 ⁇ 5 to 18:0 in the oil is between 100:1 and 1:2, and wherein the fatty acid of the oil comprises less than 10% (w/w), or less than 5%, 20: 1 A5 .
  • the present invention further provides an oil comprising fatty acids, wherein at least 43%, or at leasst 50%, or at least 60%, of the C18 fatty acid of the oil is 18: ;11 ⁇ S
  • the invention further provides an oil comprising fatty acids comprising at least 3.0% (w/w), or at least 5% (w/w) or at least 10% (w/w), 18:1 ⁇ 5 as a percentage of the total fatty acid of the oil.
  • oil comprises less than 10% (w/w), or less than 5% (w/w), 18:3 ⁇ 9 ' ⁇ 12 ' ⁇ 15 (ALA) as a percentage of the total fatty acid in the oil.
  • the present invention further provides a fatty acid produced by, or obtained from, the cell according to the invention, the transgenic non-human organism of the invention, or the seed of the invention.
  • the invention also provides a method of producing oil containing unsaturated fatty acids, the method comprising extracting oil from the cell according to the invention, the transgenic non-human organism of the invention, or the seed of the invention.
  • the present invention further provides a composition comprising a cell according to the invention, the polypeptide according to the invention, a polynucleotide according to the invention, a vector of the invention, an oil according to the invention or a fatty acid of the invention.
  • the invention also provides feedstuffs, cosmetics or chemicals comprising the oil according to the invention or a fatty acid of the invention.
  • the present invention also provides a method of performing a desaturase reaction, the method comprising contacting a substrate saturated, monounsaturated or polyunsaturated fatty acid esterified to CoA with the polypeptide of the invention
  • the present invention futher provides a substantially purified antibody, or fragment thereof, that specifically binds a polypeptide of the invention
  • the invention further provides a method of treating or preventing a condition which would benefit from a PUFA, the method comprising administering to a subject a cell according to the invention, the polypeptide according to the invention, a polynucleotide according to the invention, a vector of the invention, an oil according to the invention, a fatty acid of the invention and/or a feedstuff of the invention.
  • the condition is cardiac arrhythmia's, angioplasty, inflammation, asthma, psoriasis, osteoporosis, kidney stones, AIDS, multiple sclerosis, rheumatoid arthritis, Crohn's disease, schizophrenia, cancer, foetal alcohol syndrome, attention deficient hyperactivity disorder, cystic fibrosis, phenylketonuria, unipolar depression, aggressive hostility, adrenoleukodystophy, coronary heart disease, hypertension, diabetes, obesity, Alzheimer's disease, chronic obstructive pulmonary disease, ulcerative colitis, restenosis after angioplasty, eczema, high blood pressure, platelet aggregation, gastrointestinal bleeding, endometriosis, premenstrual syndrome, myalgic encephalomyelitis, chronic fatigue after viral infections or an ocular disease.
  • the present invention also provides use of a cell according to the invention, the polypeptide according to the invention, a polynucleotide according to the invention, a vector of the invention, an oil according the invention or a fatty acid of the invention and/or a feedstuff of the invention for the manufacture of a medicament for treating or preventing a condition which would benefit from a PUFA.
  • Figure 1 A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 2b. Dots indicate sequence identity, while different nucleotides are indicated.
  • Figure 2 A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 3. Dots indicate sequence identity, while different nucleotides are indicated.
  • Figure 3 A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 6b. Dots indicate sequence identity, while different nucleotides are indicated.
  • Figure 4 A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 10. Dots indicate sequence identity, while different nucleotides are indicated.
  • Figure 5 A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 11. Dots indicate sequence identity, while different nucleotides are indicated.
  • Figure 7 A, Gas chromatography (GC) of yeast fatty acid methyl esters from S. cerivisiae expressing AdDl 2Des.
  • B Gas chromatography (GC) of yeast fatty acid methyl esters from S. cerivisiae, vector only.
  • C Confirmation of the double bond positions of C16:2 and C18:2 products by GC-mass spectrometry.
  • Figure 8. GC analysis of yeast olel cells expressing pYES2 vector only (Panel A) or A. domesticus ⁇ 12-desaturase in pXZP282 (Panel B) after fed with mixture of C14:0 and C15:0.
  • Figure 9 Phylogenetic analysis of representative acyl-CoA or acyl-lipid desaturase protein sequences.
  • Figure 10 Phylogenetic analysis of representative acyl-CoA or acyl-lipid desaturase protein sequences.
  • SEQ ID NO: 133 coding sequence of Acheta domesticus desaturase AdDIIDes
  • SEQ ID NO: 134 amino acid sequence of Acheta domesticus desaturase AdDl 2Des
  • SEQ ID NO: 135 -Arabidopsis thaliana FAD2 ⁇ 12 desaturase
  • fatty acid refers to a carboxylic acid (or organic acid), often with a long aliphatic tail, either saturated or unsaturated.
  • fatty acids typically have a carbon-carbon bonded chain of at least 8 carbon atoms in length, more preferably at least 12 carbons in length.
  • Most naturally occurring fatty acids have an even number of carbon atoms because their biosynthesis involves acetate which has two carbon atoms.
  • the fatty acids may be in a free state (non-esterified) or in an esterified form such as part of a triglyceride, diacylglyceride, monoacylglyceride, acyl-CoA (thio-ester) bound or other bound form.
  • the fatty acid may be esterified as a phospholipid such as a phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylglycerol, phosphatidylinositol or diphosphatidylglycerol forms.
  • a phospholipid such as a phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylglycerol, phosphatidylinositol or diphosphatidylglycerol forms.
  • saturated fatty acids do not contain any double bonds or other functional groups along the chain.
  • saturated refers to hydrogen, in that all carbons (apart from the carboxylic acid [-COOH] group) contain as many hydrogens as possible.
  • the omega ( ⁇ ) end contains 3 hydrogens (CH3-) and each carbon within the chain contains 2 hydrogens (-CH2-).
  • the terms “monounsaturated fatty acid” refers to a fatty acid which comprises at least 12 carbon atoms in its carbon chain and only one alkene group in the chain.
  • the terms “polyunsaturated fatty acid” or “PUFA” refer to a fatty acid which comprises at least 12 carbon atoms in its carbon chain and at least two alkene groups (carbon-carbon double bonds).
  • the number of carbon atoms in the carbon chain of the fatty acids refers to an unbranched carbon chain. If the carbon chain is branched, the number of carbon atoms excludes those in sidegroups.
  • the long-chain polyunsaturated fatty acid is an ⁇ 3 fatty acid, that is, having a desaturation (carbon-carbon double bond) in the third carbon-carbon bond from the methyl end of the fatty acid.
  • the long-chain polyunsaturated fatty acid is an ⁇ 6 fatty acid, that is, having a desaturation (carbon-carbon double bond) in the sixth carbon-carbon bond from the methyl end of the fatty acid.
  • long-chain polyunsaturated fatty acid or "LC- PUFA” refer to a fatty acid which comprises at least 20 carbon atoms in its carbon chain and at least two carbon-carbon double bonds.
  • the term "desaturase” refers to an enzyme which is capable of introducing a carbon-carbon double bond into the acyl group of a fatty acid substrate, which is typically in an esterified form such as, for example, fatty acid CoA esters.
  • the acyl group may be esterified to a phospholipid such as phosphatidyl choline, or to acyl carrier protein (ACP), or in a preferred embodiment to CoA.
  • Desaturases generally may be categorized into three groups accordingly.
  • ⁇ 12 desaturase refers to a protein which performs a desaturase reaction that introduces a carbon-carbon double bond located at the 12 th bond from the carboxyl end and has greater activity in desaturation at this position than any other position, hi one embodiment, the ⁇ 12 desaturase activity includes oleoyl-CoA ⁇ 12 desaturase activity. In another embodiment, the ⁇ 12 desaturase activity includes palmitoleoyl-CoA ⁇ 12 desaturase activitiy. These fatty acids may be in an esterified form, such as, for example, as part of a phospholipid. Examples of ⁇ 12 desaturases include proteins comprising an amino acid sequence provided in SEQ ID NOs: 28, 78 and 134.
  • ⁇ 12 desaturases There are two types of ⁇ 12 desaturases; acyl-CoA ⁇ 12 desaturases and acyl- PC ⁇ 12 desaturases, which are predominantly active on acyl-CoA and acyl-PC linked 18:1 substrates, respectively. However, ⁇ 9 desaturases may also be active on an acyl- ACP (acyl carrier protein).
  • acyl-CoA ⁇ 12 desaturase activity or "acyl-CoA ⁇ 12 desaturase” refers to the desaturase having greater activity on an acyl-CoA substrate than an acyl-lipid (such as acyl-PC) and/or acyl-ACP substrate. In an embodiment, the activity is at least two-fold greater.
  • a " ⁇ 5 desaturase” refers to a protein which performs a desaturase reaction that introduces a carbon-carbon bond located at the 5 th bond from the carboxyl end and has greater activity in desaturation at this position than any other position, hi one embodiment the ⁇ 5 desaturase has acyl-CoA-stearoyl ⁇ 5 desaturase activity. In another embodiment, the ⁇ 5 desaturase has acyl-CoA-palmitoyl ⁇ 5 desaturase activity.
  • the enzyme ⁇ 5 desaturase catalyses the desaturation of C20 LC-PUFA, converting dihomo- ⁇ -linoleic acid DGLA to arachidonic acid (ARA, 20:4 ⁇ 6) and ETA to EPA (20:5 ⁇ 3).
  • acyl-CoA ⁇ 5 desaturase activity or "acyl-CoA ⁇ 5 desaturase” refers to the desaturase having greater activity on an acyl-CoA substrate than an acyl-lipid (such as acyl-PC) and/or acyl-ACP substrate. In an embodiment, the activity is at least two-fold greater.
  • ⁇ 9 desaturase refers to a protein which performs a desaturase reaction that introduces a carbon-carbon bond located at the 9 bond from the carboxyl end and has greater activity in desaturation at this position than any other position.
  • ⁇ 9 desaturase activity include myristoyl-CoA ⁇ 9 desaturase activity, stearoyl-CoA ⁇ 9 desaturase activity, palmitoyl-CoA ⁇ 9 desaturase activity, lignoceroyl-CoA ⁇ 9 desaturase activity and behenoyl-CoA ⁇ 9 desaturase activity.
  • acyl-CoA ⁇ 9 desaturase activity or "acyl-CoA ⁇ 9 desaturase” refers to the desaturase having greater activity on an acyl-CoA substrate than an acyl-lipid (such as acyl-PC) and/or acyl-ACP substrate. In an embodiment, the activity is at least two-fold greater.
  • conjuggase refers to a co ⁇ jutriene-forming desaturase.
  • epoxyase refers to an enzyme that introduces an epoxy group into a fatty acid resulting in the production of an epoxy fatty acid.
  • the epoxy group is introduced at the 2nd and/or 12th carbon on a fatty acid chain, especially of a C16 or C18 fatty acid chain.
  • Hydroxylase refers to an enzyme that introduces a hydroxyl group into a fatty acid resulting in the production of a hydroxylated fatty acid.
  • the hydroxyl group is introduced at the 2nd, 12th and/or 17th carbon on a Cl 8 fatty acid chain.
  • the hydroxyl group is introduced at the 15th carbon on a Cl 6 fatty acid chain.
  • plant includes whole plants, vegetative structures (for example, leaves, stems), roots, floral organs/structures, seed (including embryo, endosperm, and seed coat), plant tissue (for example, vascular tissue, ground tissue, and the like), cells and progeny of the same.
  • transgenic plant refers to a plant that contains a gene construct ("transgene") not found in a wild-type plant of the same species, variety or cultivar.
  • a “transgene” as referred to herein has the normal meaning in the art of biotechnology and includes a genetic sequence which has been produced or altered by recombinant DNA or RNA technology and which has been introduced into the plant cell.
  • the transgene may include genetic sequences derived from a plant cell.
  • the transgene has been introduced into the plant by human manipulation such as, for example, by transformation but any method can be used as one of skill in the art recognizes.
  • seed and “grain” are used interchangeably herein.
  • “Grain” generally refers to mature, harvested grain but can also refer to grain after imbibition or germination, according to the context. Mature grain commonly has a moisture content of less than about 18-20%.
  • operably linked refers to a functional relationship between two or more nucleic acid (e.g., DNA) segments. Typically, it refers to the functional relationship of transcriptional regulatory element (promoter) to a transcribed sequence.
  • a promoter is operably linked to a coding sequence, such as a polynucleotide defined herein, if it stimulates or modulates the transcription of the coding sequence in an appropriate cell.
  • promoter transcriptional regulatory elements that are operably linked to a transcribed sequence are physically contiguous to the transcribed sequence, i.e., they are cw-acting.
  • some transcriptional regulatory elements, such as enhancers need not be physically contiguous or located in close proximity to the coding sequences whose transcription they enhance.
  • the term "gene” is to be taken in its broadest context and includes the deoxyribonucleotide sequences comprising the protein coding region of a structural gene and including sequences located adjacent to the coding region on both the 5' and 3' ends for a distance of at least about 2 kb on either end and which are involved in expression of the gene.
  • the sequences which are located 5' of the coding region and which are present on the niRNA are referred to as 5' non-translated sequences.
  • the sequences which are located 3' or downstream of the coding region and which are present on the rnRNA are referred to as 3' non-translated sequences.
  • the term "gene” encompasses both cDNA and genomic forms of a gene.
  • a genomic form or clone of a gene contains the coding region which may be interrupted with non-coding sequences termed "introns” or “intervening regions” or “intervening sequences.”
  • Introns are segments of a gene which are transcribed into nuclear RNA (hnRNA); introns may contain regulatory elements such as enhancers. Introns are removed or “spliced out” from the nuclear or primary transcript; introns therefore are absent in the messenger RNA (rnRNA) transcript.
  • the mRNA functions during translation to specify the sequence or order of amino acids in a nascent polypeptide.
  • gene includes a synthetic or fusion molecule encoding all or part of the proteins of the invention described herein and a complementary nucleotide sequence to any one of the above.
  • can be isolated from means that the polynucleotide or encoded polypeptide is naturally produced by an organism, particularly an insect.
  • substantially purified polypeptide or “purified polypeptide” we mean a polypeptide that has generally been separated from the lipids, nucleic acids, other peptides, and other contaminating molecules with which it is associated in its native state.
  • the substantially purified polypeptide is at least 60% free, more preferably at least 75% free, and more preferably at least 90% free from other components with which it is naturally associated.
  • recombinant in the context of a polypeptide refers to the polypeptide when produced by a cell, or in a cell-free expression system, in an altered amount or at an altered rate compared to its native state.
  • the cell is a cell that does not naturally produce the polypeptide.
  • the cell may be a cell which comprises a non-endogenous gene that causes an altered amount of the polypeptide to be produced.
  • a recombinant polypeptide of the invention includes polypeptides which have not been separated from other components of the transgenic (recombinant) cell, or cell-free expression system, in which it is produced, and polypeptides produced in such cells or cell-free systems which are subsequently purified away from at least some other components.
  • polypeptide and “protein” are generally used interchangeably.
  • the query sequence is at least 15 amino acids in length, and the GAP analysis aligns the two sequences over a region of at least 15 amino acids. More preferably, the query sequence is at least 50 amino acids in length, and the GAP analysis aligns the two sequences over a region of at least 50 amino acids. More preferably, the query sequence is at least 100 amino acids in length and the GAP analysis aligns the two sequences over a region of at least 100 amino acids. Even more preferably, the query sequence is at least 250 amino acids in length and the GAP analysis aligns the two sequences over a region of at least 250 amino acids. Even more preferably, the GAP analysis aligns two sequences over their entire length.
  • a "biologically active" fragment is a portion of a polypeptide of the invention which maintains a defined activity of the full-length polypeptide, namely possessing desaturase, conjugase, epoxidase, and/or hydroxylase activity.
  • Biologically active fragments can be any size as long as they maintain the defined activity.
  • the biologically active fragment maintains at least 10% of the acivity of the full length protein.
  • % identity figures higher than those provided above will encompass preferred embodiments.
  • the polypeptide/enzyme comprises an amino acid sequence which is at least 60%, more preferably at least 65%, more preferably at least 70%, more preferably at least 75%, more preferably at least 76%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 91%, more preferably at least 92%, more preferably at least 93%, more preferably at least 94%, more preferably at least 95%, more preferably at least 96%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99%, more preferably at least 99.1%, more preferably at least 99.2%, more preferably at least 99.3%, more preferably at least 99.4%, more preferably at least 99.5%, more preferably at least 99.6%, more preferably at least 99.7%, more preferably at least 99.8%, and even more preferably at least 99.9% identical to the relevant nominated SEQ
  • acyl-CoA ⁇ 5 desaturases of the invention this is the first demonstration of an animal desaturase that can produce 5-hexadecenoic and 5- octadecenoic acids and uses an acyl-CoA fatty acid as a substrate.
  • acyl-CoA ⁇ 12 desaturases of the invention this is the first known demonstration of of ⁇ 12 desaturase activity by an enzyme naturally produced by an animal. Furthermore, this is the first demonstration of a desaturase that can produce linoleic acid and uses an acyl-CoA fatty acid as a substrate.
  • Amino acid sequence mutants of the polypeptides of the present invention can be prepared by introducing appropriate nucleotide changes into a nucleic acid of the present invention, or by in vitro synthesis of the desired polypeptide.
  • Such mutants include, for example, deletions, insertions or substitutions of residues within the amino acid sequence.
  • a combination of deletion, insertion and substitution can be made to arrive at the final construct, provided that the final peptide product possesses the desired characteristics.
  • Mutant (altered) peptides can be prepared using any technique known in the art.
  • a polynucleotide of the invention can be subjected to in vitro mutagenesis.
  • in vitro mutagenesis techniques include sub-cloning the polynucleotide into a suitable vector, transforming the vector into a "mutator" strain such as the E. coli XL-I red (Stratagene) and propagating the transformed bacteria for a suitable number of generations.
  • the polynucleotides of the invention are subjected to DNA shuffling techniques as broadly described by Harayama (1998). Products derived from mutated/altered DNA can readily be screened using techniques described herein to determine if they possess desaturase, conjugase, epoxidase, and/or hydroxylase activity.
  • the location of the mutation site and the nature of the mutation will depend on characteristic(s) to be modified.
  • the sites for mutation can be modified individually or in series, e.g., by (1) substituting first with conservative amino acid choices and then with more radical selections depending upon the results achieved, (2) deleting the target residue, or (3) inserting other residues adjacent to the located site.
  • Amino acid sequence deletions generally range from about 1 to 15 residues, more preferably about 1 to 10 residues and typically about 1 to 5 contiguous residues.
  • Substitution mutants have at least one amino acid residue in the polypeptide molecule removed and a different residue inserted in its place.
  • the sites of greatest interest for substitutional mutagenesis include sites identified as the active site(s).
  • Other sites of interest are those in which particular residues obtained from various strains or species are identical. These positions may be important for biological activity. These sites, especially those falling within a sequence of at least three other identically conserved sites, are preferably substituted in a relatively conservative manner. Such conservative substitutions are shown in Table 1 under the heading of "exemplary substitutions".
  • a mutant/variant polypeptide has one or two or three or four conservative amino acid changes when compared to a naturally occurring polypeptide. Details of conservative amino acid changes are provided in Table 1.
  • the changes are not in one or more of the following motifs: AGAHRLW, SETDAD, FFFSHVG, QKKY, NSAAH, GEGWHNYHH, PWDY 5 and GWAY.
  • the changes are not in each of the following motifs: AGAHRLW, SETDAD, FFFSHVG, QKKY, NSAAH, GEGWHNYHH, PWDY, and GWAY.
  • minor changes can reasonably be predicted not to alter the activity of the polypeptide when expressed in a recombinant cell.
  • unnatural amino acids or chemical amino acid analogues can be introduced as a substitution or addition into the polypeptides of the present invention.
  • Such amino acids include, but are not limited to, the D-isomers of the common amino acids, 2,4-diaminobutyric acid, ⁇ -amino isobutyric acid, 4- aminobutyric acid, 2-aminobutyric acid, 6-amino hexanoic acid, 2-amino isobutyric acid, 3 -amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, ⁇ -alanine, fluoro-amino acids, designer amino acids such as ⁇ -methyl amino acids, C ⁇ -methyl amino acids, N ⁇ -methyl
  • polypeptides of the present invention which are differentially modified during or after synthesis, e.g., by biotinylation, benzylation, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. These modifications may serve to increase the stability and/or bioactivity of the polypeptide of the invention.
  • Polypeptides of the present invention can be produced in a variety of ways, including production and recovery of natural polypeptides, production and recovery of recombinant polypeptides, and chemical synthesis of the polypeptides.
  • an isolated polypeptide of the present invention is produced by culturing a cell capable of expressing the polypeptide under conditions effective to produce the polypeptide, and recovering the polypeptide.
  • a preferred cell to culture is a recombinant cell of the present invention.
  • Effective culture conditions include, but are not limited to, effective media, bioreactor, temperature, pH and oxygen conditions that permit polypeptide production.
  • An effective medium refers to any medium in which a cell is cultured to produce a polypeptide of the present invention.
  • Such medium typically comprises an aqueous medium having assimilable carbon, nitrogen and phosphate sources, and appropriate salts, minerals, metals and other nutrients, such as vitamins.
  • Cells of the present invention can be cultured in conventional fermentation bioreactors, shake flasks, test tubes, microtiter dishes, and petri plates. Culturing can be carried out at a temperature, pH and oxygen content appropriate for a recombinant cell. Such culturing conditions are within the expertise of one of ordinary skill in the art.
  • isolated polynucleotide we mean a polynucleotide which has generally been separated from the polynucleotide sequences with which it is associated or linked in its native state.
  • the isolated polynucleotide is at least 60% free, more preferably at least 75% free, and more preferably at least 90% free from other components with which it is naturally associated.
  • polynucleotide is used interchangeably herein with the terms “nucleic acid molecule", “gene” and "mRNA”.
  • exogenous in the context of a polynucleotide refers to the polynucleotide when present in a cell, or in a cell-free expression system, in an altered amount compared to its native state.
  • the cell is a cell that does not naturally comprise the polynucleotide.
  • the cell may be a cell which comprises a non-endogenous polynucleotide resulting in an altered amount of production of the encoded polypeptide.
  • exogenous polynucleotide of the invention includes polynucleotides which have not been separated from other components of the transgenic (recombinant) cell, or cell-free expression system, in which it is present, and polynucleotides produced in such cells or cell-free systems which are subsequently purified away from at least some other components.
  • the exogenous polynucleotide (nucleic acid) can be a contiguous stretch of nucleotides existing in nature, or comprise two or more contiguous stretches of nucleotides from different sources (naturally occurring and/or synthetic) joined to form a single polynucleotide.
  • chimeric polynucleotides comprise at least an open reading frame encoding a polypeptide of the invention operably linked to a promoter suitable of driving transcription of the open reading frame in a cell of interest.
  • Polynucleotide refers to a oligonucleotide, polynucleotide or any fragment thereof. It may be DNA or RNA of genomic or synthetic origin, double-stranded or single-stranded, and combined with carbohydrate, lipids, protein, or other materials to perform a particular activity defined herein.
  • the query sequence is at least 45 nucleotides in length, and the GAP analysis aligns the two sequences over a region of at least 45 nucleotides.
  • the query sequence is at least 150 nucleotides in length, and the GAP analysis aligns the two sequences over a region of at least 150 nucleotides.
  • the query sequence is at least 300 nucleotides in length and the GAP analysis aligns the two sequences over a region of at least 300 nucleotides. Even more preferably, the GAP analysis aligns two sequences over their entire length.
  • the polynucleotide comprises a polynucleotide sequence which is at least 60%, more preferably at least 65%, more preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 91%, more preferably at least 92%, more preferably at least 93%, more preferably at least 94%, more preferably at least 95%, more preferably at least 96%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99%, more preferably at least 99.1%, more preferably at least 99.2%, more preferably at least 99.3%, more preferably at least 99.4%, more preferably at least 99.5%, more preferably at least 99.6%, more preferably at least 99.
  • a polynucleotide of the present invention may selectively hybridise to a polynucleotide that encodes a polypeptide of the present invention under stringent conditions.
  • under stringent conditions are those that (1) employ low ionic strength and high temperature for washing, for example, 0.015 M NaCl/0.0015 M sodium citrate/0.1% NaDodSO 4 at 50 0 C; (2) employ during hybridisation a denaturing agent such as formamide, for example, 50% (vol/vol) formamide with 0.1% bovine serum albumin, 0.1% Ficoll, 0.1% polyvinylpyrrolidone, 50 mM sodium phosphate buffer at pH 6.5 with 750 mM NaCl, 75 mM sodium citrate at 42 0 C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonic
  • Polynucleotides of the present invention may possess, when compared to naturally occurring molecules, one or more mutations which are deletions, insertions, or substitutions of nucleotide residues. Mutants can be either naturally occurring (that is to say, isolated from a natural source) or synthetic (for example, by performing site- directed mutagenesis or DNA shuffling on the nucleic acid as described above). It is thus apparent that polynucleotides of the invention can be either naturally occurring or recombinant.
  • Oligonucleotides of the present invention can be RNA, DNA, or derivatives of either.
  • the minimum size of such oligonucleotides is the size required for the formation of a stable hybrid between an oligonucleotide and a complementary sequence on a polynucleotide of the present invention.
  • the oligonucleotides are at least 15 nucleotides, more preferably at least 18 nucleotides, more preferably at least 19 nucleotides, more preferably at least 20 nucleotides, even more preferably at least 25 nucleotides in length.
  • the present invention includes oligonucleotides that can be used as, for example, probes to identify nucleic acid molecules, or primers to produce nucleic acid molecules. Oligonucleotides of the present invention used as a probe are typically conjugated with a label such as a radioisotope, an enzyme, biotin, a fluorescent molecule or a chemiluminescent molecule.
  • a label such as a radioisotope, an enzyme, biotin, a fluorescent molecule or a chemiluminescent molecule.
  • One embodiment of the present invention includes a recombinant vector, which comprises at least one isolated polynucleotide molecule of the present invention, inserted into any vector capable of delivering the polynucleotide molecule into a host cell.
  • a vector contains heterologous polynucleotide sequences, that is polynucleotide sequences that are not naturally found adjacent to polynucleotide molecules of the present invention and that preferably are derived from a species other than the species from which the polynucleotide molecule(s) are derived.
  • the vector can be either RNA or DNA, either prokaryotic or eukaryotic, and typically is a transposon (such as described in US 5,792,294), a virus or a plasmid.
  • One type of recombinant vector comprises a polynucleotide molecule of the present invention operatively linked to an expression vector.
  • the phrase "operably linked" refers to insertion of a polynucleotide molecule into an expression vector in a manner such that the molecule is able to be expressed when transformed into a host cell.
  • an expression vector is a DNA or RNA vector that is capable of transforming a host cell and of effecting expression of a specified polynucleotide molecule.
  • the expression vector is also capable of replicating within the host cell.
  • Expression vectors can be either prokaryotic or eukaryotic, and are typically viruses or plasmids.
  • Expression vectors of the present invention include any vectors that function (i.e., direct gene expression) in recombinant cells of the present invention, including in bacterial, fungal, endoparasite, arthropod, animal, and plant cells. Particularly preferred expression vectors of the present invention can direct gene expression in yeast and/or plants cells.
  • expression vectors of the present invention contain regulatory sequences such as transcription control sequences, translation control sequences, origins of replication, and other regulatory sequences that are compatible with the recombinant cell and that control the expression of polynucleotide molecules of the present invention.
  • recombinant molecules of the present invention include transcription control sequences. Transcription control sequences are sequences which control the initiation, elongation, and termination of transcription. Particularly important transcription control sequences are those which control transcription initiation, such as promoter, enhancer, operator and repressor sequences. Suitable transcription control sequences include any transcription control sequence that can function in at least one of the recombinant cells of the present invention. A variety of such transcription control sequences are known to those skilled in the art.
  • Particularly preferred transcription control sequences are promoters active in directing transcription in plants, either constitutively or stage and/or tissue specific, depending on the use of the plant or parts thereof.
  • These plant promoters include, but are not limited to, promoters showing constitutive expression, such as the 35S promoter of Cauliflower Mosaic Virus (CaMV), those for fruit-specific expression, such as the polygalacturonase (PG) promoter from tomato.
  • CaMV Cauliflower Mosaic Virus
  • PG polygalacturonase
  • Recombinant molecules of the present invention may also (a) contain secretory signals (i.e., signal segment nucleic acid sequences) to enable an expressed polypeptide of the present invention to be secreted from the cell that produces the polypeptide and/or (b) contain fusion sequences which lead to the expression of nucleic acid molecules of the present invention as fusion proteins.
  • suitable signal segments include any signal segment capable of directing the secretion of a polypeptide of the present invention.
  • Preferred signal segments include, but are not limited to, Nicotiana nectarin signal peptide (US 5,939,288), tobacco extensin signal, the soy oleosin oil body binding protein signal.
  • nucleic acid molecule of the present invention can be joined to a fusion segment that directs the encoded polypeptide to the proteosome, such as a ubiquitin fusion segment.
  • Recombinant molecules may also include intervening and/or untranslated sequences surrounding and/or within the nucleic acid sequences of nucleic acid molecules of the present invention.
  • Another embodiment of the present invention includes a recombinant cell comprising a host cell transformed with one or more recombinant molecules of the present invention. Transformation of a polynucleotide molecule into a cell can be accomplished by any method by which a polynucleotide molecule can be inserted into the cell. Transformation techniques include, but are not limited to, transfection, electroporation, microinjection, lipofection, adsorption, and protoplast fusion. A recombinant cell may remain unicellular or may grow into a tissue, organ or a multicellular organism.
  • Transformed polynucleotide molecules of the present invention can remain extrachromosomal or can integrate into one or more sites within a chromosome of the transformed (i.e., recombinant) cell in such a manner that their ability to be expressed is retained.
  • Suitable host cells to transform include any cell that can be transformed with a polynucleotide of the present invention.
  • Host cells of the present invention either can be endogenously (i.e., naturally) capable of producing polypeptides of the present invention or can be capable of producing such polypeptides after being transformed with at least one polynucleotide molecule of the present invention.
  • Host cells of the present invention can be any cell capable of producing at least one protein of the present invention, and include plant, bacterial, fungal (including yeast), parasite, and arthropod cells.
  • the host cell is a plant, arthropod or yeast cell.
  • Non limiting examples of arthropod cells include insect cells such as Spodoptera frugiperda (Sf) cells, e.g. Sf9, Sf21, Trichoplusia ni cells, and Drosophila S2 cells.
  • Synechococcus spp. also known as Synechocystis spp.
  • Synechococcus elongatus for example Synechococcus elongatus.
  • the cells may be of an organism suitable for a fermentation process.
  • the term the "fermentation process" refers to any fermentation process or any process comprising a fermentation step.
  • a fermentation process includes, without limitation, fermentation processes used to produce alcohols (e.g., ethanol, methanol, butanol); organic acids (e.g., citric acid, acetic acid, itaconic acid, lactic acid, gluconic acid); ketones (e.g., acetone); amino acids (e.g., glutamic acid); gases (e.g., H 2 and CO 2 ); antibiotics (e.g., penicillin and tetracycline); enzymes; vitamins (e.g., riboflavin, beta-carotene); and hormones.
  • alcohols e.g., ethanol, methanol, butanol
  • organic acids e.g., citric acid, acetic acid, itaconic acid, lactic acid, gluconic acid
  • ketones e
  • Fermentation processes also include fermentation processes used in the consumable alcohol industry (e.g., beer and wine), dairy industry (e.g., fermented dairy products), leather industry and tobacco industry.
  • Preferred fermentation processes include alcohol fermentation processes, as are well known in the art.
  • Preferred fermentation processes are anaerobic fermentation processes, as are well known in the art.
  • Suitable fermenting cells are able to ferment, i.e., convert, sugars, such as glucose or maltose, directly or indirectly into the desired fermentation product.
  • fermenting microorganisms include fungal organisms, such as yeast.
  • yeast includes Saccharomyces spp., Saccharomyces cerevisiae, Saccharomyces carlbergensis, Candida spp., Kluveromyces spp., Pichia spp., Hansenula spp., Trichoderma spp., Lipomyces starkey, and Yarrowia lipolytica.
  • yeast include strains of the Saccharomyces spp., and in particular, Saccharomyces cerevisiae.
  • Commercially available yeast include, e.g., Red Star/Lesaffre Ethanol Red (available from Red Star/Lesaffre, USA) FALI (available from Fleischmann's Yeast, a division of Burns Philp Food Inc., USA), SUPERSTART (available from Alltech), GERT STRAND (available from Gert Strand AB, Sweden) and FERMIOL (available from DSM Specialties).
  • Recombinant DNA technologies can be used to improve expression of a transformed polynucleotide molecule by manipulating, for example, the number of copies of the polynucleotide molecule within a host cell, the efficiency with which those polynucleotide molecules are transcribed, the efficiency with which the resultant transcripts are translated, and the efficiency of post-translational modifications.
  • Recombinant techniques useful for increasing the expression of polynucleotide molecules of the present invention include, but are not limited to, operatively linking polynucleotide molecules to high-copy number plasmids, integration of the polynucleotide molecule into one or more host cell chromosomes, addition of vector stability sequences to plasmids, substitutions or modifications of transcription control signals (e.g., promoters, operators, enhancers), substitutions or modifications of translational control signals (e.g., ribosome binding sites, Shine-Dalgarno sequences), modification of polynucleotide molecules of the present invention to correspond to the codon usage of the host cell, and the deletion of sequences that destabilize transcripts.
  • transcription control signals e.g., promoters, operators, enhancers
  • translational control signals e.g., ribosome binding sites, Shine-Dalgarno sequences
  • plant as used herein as a noun refers to whole plants, but as used as an adjective refers to any substance which is present in, obtained from, derived from, or related to a plant, such as for example, plant organs (e.g. leaves, stems, roots, flowers), single cells (e.g. pollen), seeds, plant cells and the like.
  • Plants provided by or contemplated for use in the practice of the present invention include both monocotyledons and dicotyledons.
  • the plants of the present invention are crop plants (for example, cereals and pulses, maize, wheat, potatoes, tapioca, rice, sorghum, millet, cassava, barley, or pea), or other legumes.
  • the plants may be grown for production of edible roots, tubers, leaves, stems, flowers or fruit.
  • the plants may be vegetables or ornamental plants.
  • the plants of the invention may be: corn (Zea mays), canola ⁇ Brassica napiis, Brassica rapa ssp.), flax (Linum usitatissimum), alfalfa (Medicago sativ ⁇ ), rice (Oryza sativa), rye (Secede cerale), sorghum ⁇ Sorghum bicolour, Sorghum vulgare), sunflower (Helianthus annus), wheat (Tritium aestivum), soybean (Glycine max), tobacco (Nicotiana tabacum), potato (Solarium tuberosum), peanuts (Arachis hypogaea), cotton (Gossypium hirsutum), sweet potato (Lopmoea batatus), cassava (Manihot esculenta), coffee (Cofea spp.), coconut
  • the plant is an angiosperm.
  • the plant is an oilseed plant, preferably an oilseed crop plant.
  • an "oilseed plant” is a plant species used for the commercial production of oils from the seeds of the plant.
  • the oilseed plant may be oil-seed rape (such as canola), maize, sunflower, soybean, sorghum, flax (linseed) or sugar beet.
  • the oilseed plant may be other Brassicas, cotton, peanut, poppy, mustard, castor bean, sesame, safflower, or nut producing plants.
  • the plant may produce high levels of oil in its fruit, such as olive, oil palm or coconut.
  • Horticultural plants to which the present invention may be applied are lettuce, endive, or vegetable brassicas including cabbage, broccoli, or cauliflower.
  • the present invention may be applied in tobacco, cucurbits, carrot, strawberry, tomato, or pepper.
  • the non-transgenic plant used to produce a transgenic plant of the invention produces oil, especially in the seed, which has less than 20%, less than 10% or less than 5% 18:2 fatty acids and/or which has less than 10% or less than 5% 18:3 fatty acids.
  • Transgenic plants as defined in the context of the present invention include plants (as well as parts and cells of said plants) and their progeny which have been genetically modified using recombinant techniques to cause production of at least one polypeptide of the present invention in the desired plant or plant organ.
  • Transgenic plants can be produced using techniques known in the art, such as those generally described in A. Slater et al., Plant Biotechnology - The Genetic Manipulation of Plants, Oxford University Press (2003), and P. Christou and H. Klee, Handbook of Plant Biotechnology, John Wiley and Sons (2004).
  • the transgenic plants are homozygous for each and every gene that has been introduced (transgene) so that their progeny do not segregate for the desired phenotype.
  • the transgenic plants may also be heterozygous for the introduced transgene(s), such as, for example, in Fl progeny which have been grown from hybrid seed. Such plants may provide advantages such as hybrid vigour, well known in the art.
  • the transgenic plants may also comprise additional transgenes involved in the production of LC-PUFAs such as, but not limited to, a ⁇ 6 desaturase, a ⁇ 9 elongase, a ⁇ 8 desaturase, a ⁇ 6 elongase, a ⁇ 5 desaturase with activity on a 20:3 substrate, an omega-desaturase, a ⁇ 9 elongase, a ⁇ 4 desaturase, a ⁇ 7 elongase and/or members of the polyketide synthase pathway.
  • additional transgenes involved in the production of LC-PUFAs such as, but not limited to, a ⁇ 6 desaturase, a ⁇ 9 elongase, a ⁇ 8 desaturase, a ⁇ 6 elongase, a ⁇ 5 desaturase with activity on a 20:3 substrate, an omega-desaturase, a ⁇ 9 e
  • a polynucleotide of the present invention may be expressed constitutively in the transgenic plants during all stages of development. Depending on the use of the plant or plant organs, the polypeptides may be expressed in a stage-specific manner. Furthermore, the polynucleotides may be expressed tissue-specifically.
  • regulatory sequences which are known or are found to cause expression of a gene encoding a polypeptide of interest in plants may be used in the present invention.
  • the choice of the regulatory sequences used depends on the target plant and/or target organ of interest.
  • Such regulatory sequences may be obtained from plants or plant viruses, or may be chemically synthesized. Such regulatory sequences are well known to those skilled in the art.
  • plant expression vectors include, for example, one or more cloned plant genes under the transcriptional control of 5' and 3' regulatory sequences and a dominant selectable marker.
  • Such plant expression vectors also can contain a promoter regulatory region (e.g., a regulatory region controlling inducible or constitutive, environmentally- or developmentally-regulated, or cell- or tissue-specific expression), a transcription initiation start site, a ribosome binding site, an RNA processing signal, a transcription termination site, and/or a polyadenylation signal.
  • a promoter regulatory region e.g., a regulatory region controlling inducible or constitutive, environmentally- or developmentally-regulated, or cell- or tissue-specific expression
  • a transcription initiation start site e.g., a regulatory region controlling inducible or constitutive, environmentally- or developmentally-regulated, or cell- or tissue-specific expression
  • a transcription initiation start site e.g., a promoter controlling inducible or constitutive, environmentally- or developmentally-regulated, or cell- or tissue-specific expression
  • Suitable promoters for constitutive expression in plants include, but are not limited to, the cauliflower mosaic virus (CaMV) 35S promoter, the Figwort mosaic virus (FMV) 35 S, the sugarcane bacilliform virus promoter, the commelina yellow mottle virus promoter, the light-inducible promoter from the small subunit of the ribulose-l,5-bis-phosphate carboxylase, the rice cytosolic triosephosphate isomerase promoter, the adenine phosphoribosyltransferase promoter of Arabidopsis, the rice actin 1 gene promoter, the mannopine synthase and octopine synthase promoters, the Adh promoter, the sucrose synthase promoter, the R gene complex promoter, and the chlorophyll ⁇ / ⁇ binding protein gene promoter.
  • CaMV cauliflower mosaic virus
  • FMV Figwort mosaic virus
  • FMV Figwort mosaic virus
  • the commelina yellow mottle virus promoter the light
  • promoters have been used to create DNA vectors that have been expressed in plants; see, e.g., PCT publication WO 8402913. All of these promoters have been used to create various types of plant- expressible recombinant DNA vectors.
  • promoters utilized in the present invention have relatively high expression in these specific tissues.
  • promoters for genes with tissue- or cell-specific or - enhanced expression. Examples of such promoters reported in the literature include the chloroplast glutamine synthetase GS2 promoter from pea, the chloroplast fructose- 1,6-biphosphatase promoter from wheat, the nuclear photosynthetic ST-LSl promoter from potato, the serine/threonine kinase promoter and the glucoamylase (CHS) promoter from Arabidopsis thaliana.
  • CHS glucoamylase
  • ribulose-l,5-bisphosphate carboxylase promoter from eastern larch (Larix laricind), the promoter for the Cab gene, Cab ⁇ , from pine, the promoter for the Cab-1 gene from wheat, the promoter for the Cab-1 gene from spinach, the promoter for the Cab IR gene from rice, the pyruvate, orthophosphate dikinase (PPDK) promoter from Zea mays, the promoter for the tobacco Lhcbl*2 gene, the Arabidopsis thaliana Suc2 sucrose-H 30 symporter promoter, and the promoter for the thylakoid membrane protein genes from spinach (PsaD, PsaF, PsaE, PC, FNR, AtpC, AtpD, Cab, RbcS).
  • promoters for the chlorophyll ⁇ / ⁇ -binding proteins may also be utilized in the present invention, such as the promoters for LhcB gene and PsbP gene from white mustard (Sinapis alba).
  • sink tissues of the plant such as the tuber of the potato plant, the fruit of tomato, or the seed of soybean, canola, cotton, Zea mays, wheat, rice, and barley, it is preferred that the promoters utilized in the present invention have relatively high expression in these specific tissues.
  • a number of promoters for genes with tuber-specific or -enhanced expression are known, including the class I patatin promoter, the promoter for the potato tuber ADPGPP genes, both the large and small subunits, the sucrose synthase promoter, the promoter for the major tuber proteins including the 22 kD protein complexes and proteinase inhibitors, the promoter for the granule bound starch synthase gene (GBSS), and other class I and II patatins promoters.
  • Other promoters can also be used to express a protein in specific tissues, such as seeds or fruits.
  • the promoter for ⁇ -conglycinin or other seed-specific promoters such as the napin and phaseolin promoters, can be used.
  • a particularly preferred promoter for Zea mays endosperm expression is the promoter for the glutelin gene from rice, more particularly the Osgt-1 promoter.
  • promoters suitable for expression in wheat include those promoters for the ADPglucose pyrosynthase (ADPGPP) subunits, the granule bound and other starch synthase, the branching and debranching enzymes, the embryogenesis-abundant proteins, the gliadins, and the glutenins.
  • ADPGPP ADPglucose pyrosynthase
  • promoters in rice include those promoters for the ADPGPP subunits, the granule bound and other starch synthase, the branching enzymes, the debranching enzymes, sucrose synthases, and the glutelins.
  • a particularly preferred promoter is the promoter for rice glutelin, Osgt- 1 gene.
  • promoters for barley include those for the ADPGPP subunits, the granule bound and other starch synthase, the branching enzymes, the debranching enzymes, sucrose synthases, the hordeins, the embryo globulins, and the aleurone specific proteins.
  • Root specific promoters may also be used.
  • An example of such a promoter is the promoter for the acid chitinase gene. Expression in root tissue could also be accomplished by utilizing the root specific subdomains of the CaMV 35 S promoter that have been identified.
  • the promoter directs expression in tissues and organs in which fatty acid and oil biosynthesis take place, particularly in seed cells such as endosperm cells and cells of the developing embryo.
  • Promoters which are suitable are the oilseed rape napin gene promoter (US 5,608,152), the Vicia faba USP promoter (Baumlein et al, 1991), the Arabidopsis oleosin promoter (WO 98/45461), the Phaseolus vulgaris phaseolin promoter (US 5,504,200), the Brassica Bce4 promoter (WO 91/13980) or the legumin B4 promoter (Baumlein et al., 1992), and promoters which lead to the seed-specific expression in monocots such as maize, barley, wheat, rye, rice and the like.
  • promoters which are suitable are the barley Ipt2 or lptl gene promoter (WO 95/15389 and WO 95/23230) or the promoters described in WO 99/16890 (promoters from the barley hordein gene, the rice glutelin gene, the rice oryzin gene, the rice prolamin gene, the wheat gliadin gene, the wheat glutelin gene, the maize zein gene, the oat glutelin gene, the sorghum kasirin gene, the rye secalin gene).
  • Other promoters include those described by Broun et al. (1998) and US 20030159173.
  • the 5' non-translated leader sequence can be derived from the promoter selected to express the heterologous gene sequence of the polynucleotide of the present invention, and can be specifically modified if desired so as to increase translation of mRNA.
  • the 5' non-translated regions can also be obtained from plant viral RNAs (Tobacco mosaic virus, Tobacco etch virus, Maize dwarf mosaic virus, Alfalfa mosaic virus, among others) from suitable eukaryotic genes, plant genes (wheat and maize chlorophyll a/b binding protein gene leader), or from a synthetic gene sequence.
  • the present invention is not limited to constructs wherein the non- translated region is derived from the 5' non-translated sequence that accompanies the promoter sequence.
  • the leader sequence could also be derived from an unrelated promoter or coding sequence.
  • Leader sequences useful in context of the present invention comprise the maize Hsp70 leader (U.S. 5,362,865 and U.S. 5,859,347), and the TMV omega element.
  • the termination of transcription is accomplished by a 3' non-translated DNA sequence operably linked in the chimeric vector to the polynucleotide of interest.
  • the 3' non-translated region of a recombinant DNA molecule contains a polyadenylation signal that functions in plants to cause the addition of adenylate nucleotides to the 3' end of the RNA.
  • the 3' non-translated region can be obtained from various genes that are expressed in plant cells.
  • the nopaline synthase 3' untranslated region, the 3' untranslated region from pea small subunit Rubisco gene, the 3' untranslated region from soybean 7S seed storage protein gene are commonly used in this capacity.
  • the 3' transcribed, non-translated regions containing the polyadenylate signal of Agrobacterium tumor-inducing (Ti) plasmid genes are also suitable.
  • Acceleration methods include, for example, microprojectile bombardment and the like.
  • microprojectile bombardment One example of a method for delivering transforming nucleic acid molecules to plant cells is microprojectile bombardment. This method has been reviewed by Yang et al., Particle Bombardment Technology for Gene Transfer, Oxford Press, Oxford, England (1994).
  • Non-biological particles that may be coated with nucleic acids and delivered into cells by a propelling force.
  • Exemplary particles include those comprised of tungsten, gold, platinum, and the like.
  • An illustrative embodiment of a method for delivering DNA into Zea mays cells by acceleration is a biolistics ⁇ -particle delivery system, that can be used to propel particles coated with DNA through a screen, such as a stainless steel or Nytex screen, onto a filter surface covered with corn cells cultured in suspension.
  • a particle delivery system suitable for use with the present invention is the helium acceleration PDS- 1000/He gun available from Bio-Rad Laboratories.
  • cells in suspension may be concentrated on filters.
  • Filters containing the cells to be bombarded are positioned at an appropriate distance below the microprojectile stopping plate. If desired, one or more screens are also positioned between the gun and the cells to be bombarded.
  • immature embryos or other target cells may be arranged on solid culture medium.
  • the cells to be bombarded are positioned at an appropriate distance below the microprojectile stopping plate.
  • one or more screens are also positioned between the acceleration device and the cells to be bombarded.
  • bombardment transformation one may optimize the pre-bombardment culturing conditions and the bombardment parameters to yield the maximum numbers of stable transformants.
  • Both the physical and biological parameters for bombardment are important in this technology. Physical factors are those that involve manipulating the DNA/microprojectile precipitate or those that affect the flight and velocity of either the macro- or microprojectiles.
  • Biological factors include all steps involved in manipulation of cells before and immediately after bombardment, the osmotic adjustment of target cells to help alleviate the trauma associated with bombardment, and also the nature of the transforming DNA 5 such as linearized DNA or intact supercoiled plasmids. It is believed that pre-bombardment manipulations are especially important for successful transformation of immature embryos.
  • plastids can be stably transformed.
  • Methods disclosed for plastid transformation in higher plants include particle gun delivery of DNA containing a selectable marker and targeting of the DNA to the plastid genome through homologous recombination (U.S. 5, 451,513, U.S. 5,545,818, U.S. 5,877,402, U.S. 5,932479, and WO 99/05265).
  • the execution of other routine adjustments will be known to those of skill in the art in light of the present disclosure.
  • Agrobacterium-medi&ted transfer is a widely applicable system for introducing genes into plant cells because the DNA can be introduced into whole plant tissues, thereby bypassing the need for regeneration of an intact plant from a protoplast.
  • the use of Agrobacterium-mediatQd plant integrating vectors to introduce DNA into plant cells is well known in the art (see, for example, US 5,177,010, US 5,104,310, US 5,004,863, US 5,159,135). Further, the integration of the T-DNA is a relatively precise process resulting in few rearrangements.
  • the region of DNA to be transferred is defined by the border sequences, and intervening DNA is usually inserted into the plant genome.
  • Modern Agrobacterium transformation vectors are capable of replication in E. coli as well as Agrobacterium, allowing for convenient manipulations as described (Klee et al., In: Plant DNA Infectious Agents, Hohn and Schell, eds., Springer-Verlag, New York, pp. 179-203 (1985).
  • technological advances in vectors for Agrobacterium-mediaXed gene transfer have improved the arrangement of genes and restriction sites in the vectors to facilitate construction of vectors capable of expressing various polypeptide coding genes.
  • the vectors described have convenient multi-linker regions flanked by a promoter and a polyadenylation site for direct expression of inserted polypeptide coding genes and are suitable for present purposes.
  • Agrobacterium containing both armed and disarmed Ti genes can be used for the transformations. In those plant varieties where Agrobacterium-medistQd transformation is efficient, it is the method of choice because of the facile and defined nature of the gene transfer.
  • a transgenic plant formed using Agrohacterium transformation methods typically contains a single genetic locus on one chromosome. Such transgenic plants can be referred to as being hemizygous for the added gene. More preferred is a transgenic plant that is homozygous for the added gene; i.e., a transgenic plant that contains two added genes, one gene at the same locus on each chromosome of a chromosome pair.
  • a homozygous transgenic plant can be obtained by sexually mating (selfing) an independent segregant transgenic plant that contains a single added gene, germinating some of the seed produced and analyzing the resulting plants for the gene of interest.
  • transgenic plants can also be mated to produce offspring that contain two independently segregating exogenous genes. Selfing of appropriate progeny can produce plants that are homozygous for both exogenous genes.
  • Back-crossing to a parental plant and out-crossing with a non- transgenic plant are also contemplated, as is vegetative propagation. Descriptions of other breeding methods that are commonly used for different traits and crops can be found in Fehr, In: Breeding Methods for Cultivar Development, Wilcox J. ed., American Society of Agronomy, Madison Wis. (1987).
  • Transformation of plant protoplasts can be achieved using methods based on calcium phosphate precipitation, polyethylene glycol treatment, electroporation, and combinations of these treatments. Application of these systems to different plant varieties depends upon the ability to regenerate that particular plant strain from protoplasts. Illustrative methods for the regeneration of cereals from protoplasts are described (Fujimura et al, 1985; Toriyama et al, 1986; Abdullah et al., 1986).
  • Other methods of cell transformation can also be used and include but are not limited to introduction of DNA into plants by direct DNA transfer into pollen, by direct injection of DNA into reproductive organs of a plant, or by direct injection of DNA into the cells of immature embryos followed by the rehydration of desiccated embryos.
  • the regeneration, development, and cultivation of plants from single plant protoplast transformants or from various transformed explants is well known in the art (Weissbach et al., In: Methods for Plant Molecular Biology, Academic Press, San Diego, Calif., (1988).
  • This regeneration and growth process typically includes the steps of selection of transformed cells, culturing those individualized cells through the usual stages of embryonic development through the rooted plantlet stage. Transgenic embryos and seeds are similarly regenerated. The resulting transgenic rooted shoots are thereafter planted in an appropriate plant growth medium such as soil.
  • the development or regeneration of plants containing the foreign, exogenous gene is well known in the art.
  • the regenerated plants are self-pollinated to provide homozygous transgenic plants.
  • a transgenic plant of the present invention containing a desired exogenous nucleic acid is cultivated using methods well known to one skilled in the art.
  • transgenic wheat or barley plants are produced by Agrobacterium tumefaciens mediated transformation procedures.
  • Vectors carrying the desired nucleic acid construct may be introduced into regenerable wheat cells of tissue cultured plants or explants, or suitable plant systems such as protoplasts.
  • the regenerable wheat cells are preferably from the scutellum of immature embryos, mature embryos, callus derived from these, or the meristematic tissue.
  • transgenic plants can be grown to produce plant tissues or parts having the desired phenotype.
  • the plant tissue or plant parts may be harvested, and/or the seed collected. The seed may serve as a source for growing additional plants with tissues or parts having the desired characteristics.
  • transgenic non-human animal refers to an animal, other than a human, that contains a gene construct ("transgene") not found in a wild-type animal of the same species or breed.
  • a "transgene” as referred to herein has the normal meaning in the art of biotechnology and includes a genetic sequence which has been produced or altered by recombinant DNA or RNA technology and which has been introduced into an animal cell.
  • the transgene may include genetic sequences derived from an animal cell.
  • the transgene has been introduced into the animal by human manipulation such as, for example, by transformation but any method can be used as one of skill in the art recognizes.
  • Heterologous DNA can be introduced, for example, into fertilized mammalian ova.
  • totipotent or pluripotent stem cells can be transformed by microinjection, calcium phosphate mediated precipitation, liposome fusion, retroviral infection or other means, the transformed cells are then introduced into the embryo, and the embryo then develops into a transgenic animal.
  • developing embryos are infected with a retrovirus containing the desired DNA, and transgenic animals produced from the infected embryo.
  • the appropriate DNAs are coinjected into the pronucleus or cytoplasm of embryos, preferably at the single cell stage, and the embryos allowed to develop into mature transgenic animals.
  • Another method used to produce a transgenic animal involves microinjecting a nucleic acid into pro-nuclear stage eggs by standard methods. Injected eggs are then cultured before transfer into the oviducts of pseudopregnant recipients.
  • Transgenic animals may also be produced by nuclear transfer technology. Using this method, fibroblasts from donor animals are stably transfected with a plasmid incorporating the coding sequences for a binding domain or binding partner of interest under the control of regulatory sequences. Stable transfectants are then fused to enucleated oocytes, cultured and transferred into female recipients.
  • feedstuffs include any food or preparation for human or animal consumption (including for enteral and/or parenteral consumption) which when taken into the body (a) serve to nourish or build up tissues or supply energy; and/or (b) maintain, restore or support adequate nutritional status or metabolic function.
  • Feedstuffs of the invention include nutritional compositions for babies and/or young children.
  • Feedstuffs of the invention comprise, for example, a cell of the invention, a plant of the invention, the plant part of the invention, the seed of the invention, an extract of the invention, the product of the method of the invention, the product of the fermentation process of the invention, or a composition along with a suitable carrier(s).
  • carrier is used in its broadest sense to encompass any component which may or may not have nutritional value. As the skilled addressee will appreciate, the carrier must be suitable for use (or used in a sufficiently low concentration) in a feedstuff such that it does not have deleterious effect on an organism which consumes the feedstuff.
  • the feedstuff of the present invention comprises an oil, fatty acid ester, or fatty acid produced directly or indirectly by use of the methods, cells or plants disclosed herein.
  • the composition may either be in a solid or liquid form. Additionally, the composition may include edible macronutrients, vitamins, and/or minerals in amounts desired for a particular use. The amounts of these ingredients will vary depending on whether the composition is intended for use with normal individuals or for use with individuals having specialized needs, such as individuals suffering from metabolic disorders and the like.
  • suitable carriers with nutritional value include, but are not limited to, macronutrients such as edible fats, carbohydrates and proteins.
  • edible fats include, but are not limited to, coconut oil, borage oil, fungal oil, black current oil, soy oil, and mono- and diglycerides.
  • carbohydrates include (but are not limited to): glucose, edible lactose, and hydrolyzed search.
  • proteins which may be utilized in the nutritional composition of the invention include (but are not limited to) soy proteins, electrodialysed whey, electrodialysed skim milk, milk whey, or the hydrolysates of these proteins.
  • vitamins and minerals may be added to the feedstuff compositions of the present invention: calcium, phosphorus, potassium, sodium, chloride, magnesium, manganese, iron, copper, zinc, selenium, iodine, and Vitamins A, E, D, C, and the B complex. Other such vitamins and minerals may also be added.
  • the components utilized in the feedstuff compositions of the present invention can be of semi-purified or purified origin.
  • semi-purified or purified is meant a material which has been prepared by purification of a natural material or by de novo synthesis.
  • a feedstuff composition of the present invention may also be added to food even when supplementation of the diet is not required.
  • the composition may be added to food of any type, including (but not limited to): margarine, modified butter, cheeses, milk, yogurt, chocolate, candy, snacks, salad oils, cooking oils, cooking fats, meats, fish and beverages.
  • the genus Saccharomyces spp is used in both brewing of beer and wine making and also as an agent in baking, particularly bread.
  • Yeast is a major constituent of vegetable extracts. Yeast is also used as an additive in animal feed. It will be apparent that genetically engineered yeast strains can be provided which are adapted to synthesise LC-PUFA as described herein. These yeast strains can then be used in food stuffs and in wine and beer making to provide products which have enhanced fatty acid content.
  • fatty acids produced in accordance with the present invention or host cells transformed to contain and express the subject genes may also be used as animal food supplements to alter an animal's tissue or milk fatty acid composition to one more desirable for human or animal consumption. Examples of such animals include sheep, cattle, horses and the like.
  • feedstuffs of the invention can be used in aquaculture to increase the levels of fatty acids in fish for human or animal consumption.
  • compositions particularly pharmaceutical compositions, comprising one or more of the fatty acids and/or resulting oils produced using the methods of the invention.
  • a pharmaceutical composition may comprise one or more of the fatty acids and/or oils, in combination with a standard, well-known, non-toxic pharmaceutically- acceptable carrier, adjuvant or vehicle such as phosphate-buffered saline, water, ethanol, polyols, vegetable oils, a wetting agent or an emulsion such as a water/oil emulsion.
  • the composition may be in either a liquid or solid form.
  • the composition may be in the form of a tablet, capsule, ingestible liquid or powder, injectible, or topical ointment or cream. Proper fluidity can be maintained, for example, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • composition can also include isotonic agents, for example, sugars, sodium chloride, and the like.
  • isotonic agents for example, sugars, sodium chloride, and the like.
  • the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfuming agents.
  • Suspensions in addition to the active compounds, may comprise suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth or mixtures of these substances.
  • suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth or mixtures of these substances.
  • Solid dosage forms such as tablets and capsules can be prepared using techniques well known in the art.
  • fatty acids produced in accordance with the present invention can be tableted with conventional tablet bases such as lactose, sucrose, and cornstarch in combination with binders such as acacia, cornstarch or gelatin, disintegrating agents such as potato starch or alginic acid, and a lubricant such as stearic acid or magnesium stearate.
  • Capsules can be prepared by incorporating these excipients into a gelatin capsule along with antioxidants and the relevant fatty acid(s).
  • the fatty acids produced in accordance with the present invention or derivatives thereof may be incorporated into commercial formulations.
  • a typical dosage of a particular fatty acid is from 0.1 mg to 20 g, taken from one to five times per day (up to 100 g daily) and is preferably in the range of from about 10 mg to about 1, 2, 5, or 10 g daily (taken in one or multiple doses).
  • any amount of fatty acid will be beneficial to the subject.
  • Possible routes of administration of the pharmaceutical compositions of the present invention include, for example, enteral (e.g., oral and rectal) and parenteral.
  • enteral e.g., oral and rectal
  • parenteral e.g., a liquid preparation may be administered orally or rectally.
  • a homogenous mixture can be completely dispersed in water, admixed under sterile conditions with physiologically acceptable diluents, preservatives, buffers or propellants to form a spray or inhalant.
  • the dosage of the composition to be administered to the patient may be determined by one of ordinary skill in the art and depends upon various factors such as weight of the patient, age of the patient, overall health of the patient, past history of the patient, immune status of the patient, etc.
  • compositions of the present invention may be utilized for cosmetic purposes. It may be added to pre-existing cosmetic compositions such that a mixture is formed or a fatty acid produced according to the subject invention may be used as the sole "active" ingredient in a cosmetic composition.
  • oilseeds can be tempered by spraying them with water to raise the moisture content to, e.g., 8.5%, and flaked using a smooth roller with a gap setting of 0.23 to 0.27 mm.
  • water may not be added prior to crushing.
  • Heat deactivates enzymes, facilitates further cell rupturing, coalesces the oil droplets, and agglomerates protein particles, all of which facilitate the extraction process.
  • the majority of the seed oil is released by passage through a screw press. Cakes expelled from the screw press are then solvent extracted, e.g., with hexane, using a heat traced column.
  • crude oil produced by the pressing operation can be passed through a settling tank with a slotted wire drainage top to remove the solids that are expressed with the oil during the pressing operation.
  • the clarified oil can be passed through a plate and frame filter to remove any remaining fine solid particles. If desired, the oil recovered from the extraction process can be combined with the clarified oil to produce a blended crude oil.
  • Degumming can be performed by addition of concentrated phosphoric acid to the crude oil to convert non- hydratable phosphatides to a hydratable form, and to chelate minor metals that are present. Gum is separated from the oil by centrifugation. The oil can be refined by addition of a sufficient amount of a sodium hydroxide solution to titrate all of the fatty acids and removing the soaps thus formed.
  • Deodorization can be performed by heating the oil to 26O 0 C under vacuum, and slowly introducing steam into the oil at a rate of about 0.1 ml/minute/100 ml of oil. After about 30 minutes of sparging, the oil is allowed to cool under vacuum. The oil is typically transferred to a glass container and flushed with argon before being stored under refrigeration. If the amount of oil is limited, the oil can be placed under vacuum, e.g., in a Parr reactor and heated to 26O 0 C for the same length of time that it would have been deodorized. This treatment improves the color of the oil and removes a majority of the volatile substances.
  • the invention also provides monoclonal or polyclonal antibodies to polypeptides of the invention or fragments thereof.
  • the present invention further provides a process for the production of monoclonal or polyclonal antibodies to polypeptides of the invention.
  • the term "binds specifically” refers to the ability of the antibody to bind to proteins of the present invention but not other known desaturase, conjugase, epoxidase and/or hydroxylase-like polypeptides.
  • epitope refers to a region of a polypeptide of the invention which is bound by the antibody.
  • An epitope can be administered to an animal to generate antibodies against the epitope, however, antibodies of the present invention preferably specifically bind the epitope region in the context of the entire polypeptide.
  • polyclonal antibodies are desired, a selected mammal (e.g., mouse, rabbit, goat, horse, etc.) is immunised with an immunogenic polypeptide such as those provided as SEQ ID NOs: 16 to 30, 73 to 78, 80 and 134. Serum from the immunised animal is collected and treated according to known procedures. If serum containing polyclonal antibodies contains antibodies to other antigens, the polyclonal antibodies can be purified by immunoaffinity chromatography. Techniques for producing and processing polyclonal antisera are known in the art. In order that such antibodies may be made, the invention also provides peptides of the invention or fragments thereof haptenised to another peptide for use as immunogens in animals.
  • an immunogenic polypeptide such as those provided as SEQ ID NOs: 16 to 30, 73 to 78, 80 and 134. Serum from the immunised animal is collected and treated according to known procedures. If serum containing polyclonal antibodies contains antibodies to other antigens, the polyclonal antibodies can
  • Monoclonal antibodies directed against polypeptides of the invention can also be readily produced by one skilled in the art.
  • the general methodology for making monoclonal antibodies by hybridomas is well known.
  • Immortal antibody-producing cell lines can be created by cell fusion, and also by other techniques such as direct transformation of B lymphocytes with oncogenic DNA, or transfection with Epstein- Barr virus.
  • Panels of monoclonal antibodies produced can be screened for various properties; i.e., for isotype and epitope affinity.
  • An alternative technique involves screening phage display libraries where, for example the phage express scFv fragments on the surface of their coat with a large variety of complementarity determining regions (CDRs). This technique is well known in the art.
  • the term "antibody”, unless specified to the contrary, includes fragments of whole antibodies which retain their binding activity for a target antigen. Such fragments include Fv, F(ab') and F(ab') 2 fragments, as well as single chain antibodies (scFv). Furthermore, the antibodies and fragments thereof may be humanised antibodies, for example as described in EP-A-239400.
  • Antibodies of the invention may be bound to a solid support and/or packaged into kits in a suitable container along with suitable reagents, controls, instructions and the like.
  • antibodies of the present invention are detectably labeled.
  • Exemplary detectable labels that allow for direct measurement of antibody binding include radiolabels, fluorophores, dyes, magnetic beads, chemiluminescers, colloidal particles, and the like.
  • Examples of labels which permit indirect measurement of binding include enzymes where the substrate may provide for a coloured or fluorescent product.
  • Additional exemplary detectable labels include covalently bound enzymes capable of providing a detectable product signal after addition of suitable substrate.
  • suitable enzymes for use in conjugates include horseradish peroxidase, alkaline phosphatase, malate dehydrogenase and the like. Where not commercially available, such antibody-enzyme conjugates are readily produced by techniques known to those skilled in the art.
  • detectable labels include biotin, which binds with high affinity to avidin or streptavidin; fluorochromes (e.g., phycobiliproteins, phycoerythrin and allophycocyanins; fluorescein and Texas red), which can be used with a fluorescence activated cell sorter; haptens; and the like.
  • the detectable label allows for direct measurement in a plate luminometer, e.g., biotin.
  • Such labeled antibodies can be used in techniques known in the art to detect polypeptides of the invention.
  • Tribdesatl (coding sequence provided as SEQ ID NO:1, amino acid sequence provided as SEQ ID NO:16), Tribdesat2a (coding sequence provided as SEQ ID NO:2, amino acid sequence provided as SEQ ID NO: 17), Tribdesat2b (coding sequence provided as SEQ ID NO:3, amino acid sequence provided as SEQ ID NO: 18), Tribdesat2c (coding sequence provided as SEQ ID NO:4, amino acid sequence provided as SEQ ID NO: 19), Tribdesat3 (coding sequence provided as SEQ ID NO:5, amino acid sequence provided as SEQ ID NO:20), Tribdesat4 (coding sequence provided as SEQ ID NO: 6, amino acid sequence provided as SEQ ID NO:21), Tribdesat5 (coding sequence provided as SEQ ID NO: 7, amino acid sequence provided as SEQ ID NO:22), Tribdesat ⁇ a (coding sequence provided as SEQ ID NO:8, amino acid sequence provided as SEQ ID NO:23), Tribdesat ⁇ b
  • His boxes are motifs that are strongly conserved in all desaturases.
  • the histidine residues in these boxes are thought to be involved in binding iron atoms required for desaturase activity.
  • the amino acid positions and sequences of the His boxes in the proteins and two others from Chauliognathus lugubris are listed in Table 3, after correction of the predicted protein sequences from analysis of the cDNA clones (Examples 2-5).
  • Concensus amino acid sequences of motifs 1, AGAPIRLW (SEQ ID NO: 104); 2, SETDAD (SEQ ID NO: 105); 3, FFFSHVG (SEQ ID NO: 106); 4, QKKY (SEQ ID NO: 107); 5, NSAAH (SEQ ID NO: 108); 6, GEGWHNYHH (SEQ ID NO: 109); 7, PWDY (SEQ ID NO: 110); 8, GWAY (SEQ ID NO: 111).
  • Table 3 Amino acid location and sequences of histidine boxes in Triboliwn and soldier beetle desaturase sequences. The number for each box corresponds to the location in the protein of the first amino acid residue of the motif.
  • Tribdesat4 using inverse PCR on cDNA as we could not predict from the genome sequence either the beginning or the end of the gene.
  • the ligation was concentrated by glycogen precipitation (150 ⁇ g glycogen, 1.4 ml ice-cold 95% ethanol at -80oC for 2 hours) after which DNA was pelleted by centrifugation (12 000g, 20 min). The DNA was resuspended in lO ⁇ l sterile water.
  • the inverse PCR reaction was performed on 1 ⁇ l of this precipitated DNA and included 5 ⁇ l 1OX Advantage 2 PCR Buffer (Clontech), 10 pmol Desat4 GSPl (5'ATTATGAGCGGATCGGCTTCCAAGTC (SEQ ID NO: 35), 10 pmol Desat4 GSP2 (5'CGAAACAATTTGGAATTCGTTTTGGG (SEQ ID NO: 36), 200 ⁇ M each dNTP, l ⁇ l 5OX BD Advantage 2 Polymerase and sterile water to a final volume of 50 ⁇ l.
  • the PCR conditions were as follows: 95oC/l min, then 30 cycles of 95oC/30 sec, 68oC/3 min, then 68oC/3 min.
  • Tribdesat4 An aliquot (5 ⁇ l) of the PCR was separated on a 1% agarose gel. A single band was noted and cloned into pGEM-T-Easy. The insert was sequenced and shown to contain most of the 5' end of Tribdesat4 and the entire 3' end of Tribdesat4. The existing 5' end of Tribdesat4 was then subject to BLASTN analysis against available sequences of ESTs from T. castaneum. One EST was identified from this analysis (Accession No. DT795463) and the rest of the 5' end was identified.
  • RNA was used in an RT-PCR reaction to obtain full length Tribdesat4.
  • the RT-PCR reaction consisted of 25 ⁇ l 2X Reaction mix (Invitrogen), 37 ng RNA, 1 ⁇ l Superscript II RT/Platinum Taq DNA polymerase (Invitrogen), 100 pmol of each primer and sterile water to 50 ⁇ l.
  • the reaction conditions were as follows: 50oC/30 min, 94oC/2 min, then 35 cycles of 94oC/30 sec, 55oC/l min, 72oC/l min, then 72oC/5 min. An aliquot was run on a 1% agarose gel.
  • SEQ ID NO: 6 shows the nucleotide sequence of the coding region of the cDNA.
  • RNA was pelleted by centrifugation at 12,000g/10 minutes at 4°C. The RNA pellet was washed once with 75% ethanol and then air-dried for 10 minutes. The RNA was then dissolved in 30 ⁇ l of RNase-free water. There was a noticeable brown colouration to the RNA which was further purified using the RNeasy mini protocol for RNA cleanup (QIAgen), according to the manufacturer's instructions.
  • RT-PCR amplification reactions were carried out for 10 desaturase sequences (Tribdesatl, 2a, 2b, 3, 5, 6a, 6b, 8, 10 and 11).
  • To each sample was added 25 ⁇ l 2X Reaction mix (Invitrogen), 37 ng RNA, 1 ⁇ l Superscript II RT/Platinum Taq DNA polymerase (Invitrogen), 100 pmol of each primer and sterile water to 50 ⁇ l.
  • the reaction conditions were as follows: 50°C/30 min, 94°C/2 min, then 35 cycles of 94°C/30 sec, 55°C/1 min, 72°C/1 min, then 72°C/5 min (PCR conditions 55/72).
  • RT-PCR reactions were carried out to amplify 6 desaturase sequences (Tribdesat2a, 2b, 2c, 6b, 8 and 12) by the method see as above. No amplification products were observed after a first round of PCR and so the reactions were subject to a second round of PCR. Amplification products of about 1 kb were observed in the Tribdesat2b, Tribdesat2c, Tribdesatl2 and Tribdesat8 samples. These products were extracted from an agarose gel using the QIAquick gel extraction kit (QIAgen) and cloned into pGEM T Easy (Promega). The DNA sequences of clones containing inserts were examined. Comparisons of gene predictions with those of RT-PCR products obtained for Tribdesat2b and 6b are shown in Figures 1 and 3, respectively.
  • PCR reactions were setup containing 1 ⁇ l dNTPs (200 ⁇ M each dNTP), 5 ⁇ l 10x ThermoPol Buffer (NEB), 1.5 ⁇ l F2 (5 ⁇ TTCTTCTTCKCNCAYKTHGGNTGG (SEQ ID NO: 82)), 1.5 ⁇ l R2 (5TGRTGGTAGTTGTGVHANCCCTC (SEQ ID NO: 83)), 5 ⁇ l cDNA, 0.1 U Taq DNA Polymerase (NEB) and sterile water to 50 ⁇ l.
  • the PCR conditions were as follows: an initial denaturation of 94 0 C for 3 minutes and then 30 cycles of 94°C/15s, 48°C/30s, 72°C/2 min and then a final extension of 72°C/5 min. An aliquot was separated on a 1.5% agarose gel and a band of approximately 400 bp was visualised.
  • the PCR reaction was purified using the QIAgen QIAquick PCR purification kit according to the manufacturer's instructions and cloned into pGEM T Easy (Promgea). Approximately 30 clones containing inserts were examined by DNA sequence analysis. A total of three unique desaturase sequences were identified corresponding to CL6 (SEQ ID NO: 75), CL7 (SEQ ID NO: 76) and CL8 (SEQ ID NO: 77).
  • RNA was extracted and cDNA synthesised as described above.
  • Two degenerate PCRs were performed on female adult RNA using the degenerate primers F2/R2 and Clu_f (5OCNCAYMGNYTNTGGGCNCA (SEQ ID NO: 84)) / Clu-r (5 ⁇ AANRYRTGRTGGTAGTTGIG (SEQ ID NO: 85)) (see Table 4) by the same method as above.
  • Amplification products of approximately 400 bp (F2/R2) and 550 bp (Clu_f/Clu_r) were visualised and cloned into pGEM T Easy.
  • Each insert from 70 colonies from the F2/R2 transformation was amplified by PCR using the T7 (5 ' TAATACGACTCACTATAGGG (SEQ ID NO: 86)) and SP6 (5 ⁇ ATTTAGGTGACACTATAG (SEQ ID NO: 87)) primers and analysed. Since approximately 70% of the sequenced clones from male soldier beetle tissue that had sequence similarity with desaturases were CL6, the PCR products were digested with restriction enzyme Rsal to remove CL6 clones. Of the 70 PCR products obtained, only 20 were not digested with Rsal. Resultant desaturase fragments obtained were CL7, CL8 and a new unique desaturase fragment called CL9.
  • Amplification of degenerate PCR from defence gland tissue A mixed population of males and females were dissected to obtain their defence gland tissue. Initially, the heads and the elytra were removed. The remaining tissue was divided into two parts - the abdomen sample and the defence gland tissue.
  • PCR products obtained were purified using the QIAgen QIAquick PCR purification kit and cloned into pGEM-T-Easy. Eleven clones were examined by sequence analysis. New desaturase gene fragments were obtained and designated CL3 and CL5. The CL5 internal fragment appeared to contain remnants of an intron, as determined by a section that did not show close sequence similarity with desaturases and also affected the reading frame, thereby inserting a stop codon.
  • the Clontech CreatorTM SMARTTM System was used to obtain both 5 ⁇ and 3 ⁇ RACE products according to the manufacturer's instructions with RNA from either abdomen or defence gland tissue.
  • Oligonucleotide primers Smart IV (5 ⁇ AAGCAGTGGTATCAACGCAGAGTGGCCATTACGGCCGGG (SEQ ID NO: 97)
  • the 5' RACE reactions used primer CDS V 5"AAGCAGTGGTATCAACGCAGAGT (SEQ ID NO: 99), and CLXfront primer.
  • the 3" RACE reactions used primers CDS III/3" PCR and CLXend.
  • Clones were screened by colony PCR for insertions. Any clones that gave a band of the expected size ( ⁇ 350 bp) were examined by DNA sequence analysis. The DNA sequence was initially examined for sequence similarity with desaturases. Full length sequences were compiled in silico using sequence information from internal sequence, 5 ⁇ ACE and 3'RACE products and examined for an open reading frame beginning with an ATG and ending in a stop codon that when translated possessed all the motifs of desaturases (Table 2).
  • Full length cDNA clones of CLl and other desaturase-like sequences were obtained using a one-step RT-PCR kit from Invitrogen according to the manufacturer's instructions and PCR conditions 55/72 (above).
  • the primers used were GGTACCATGCCACCCAACGCCCAC (SEQ ID NO: 100) and GAATTCTCACTTTTGTTTGTGAGT (SEQ ID NO: 101). Bands of approximately 1 kb were purified using the QIAquick PCR purification kit from QIAgen according to the manufacturer's instructions and cloned into pGEM-T-Easy. Positive clones were obtained and examined by DNA sequence analysis.
  • the pGEM-T-Easy clones containing the desaturase cDNAs were digested with restriction enzymes to excise the inserts, separated on a 1% agarose gel and the ⁇ lkb fragments were excised and purified using the QIAquick gel extraction kit (QIAgen) for cloning into yeast expression vectors such as pYES2 (below).
  • Chaulioenathus nobiliatus is another species of soldier beetle, which resembles C. lugubris but is generally smaller in size and less prevalent in the south-east regions of Australia. These beetle species are not closely related to moths such as Tribolium, indeed they are widely separated within the Insectae.
  • QIAquick PCR purification kit QIAquick PCR purification kit
  • a second approach used to isolate internal desaturase fragments from C. nobiliatus was to use primers specific to C. lugubris desaturases.
  • the cDNA synthesised as described above was used as a template for PCR with specific primers to CL6, CL7, CL9, and CLl.
  • An aliquot of each reaction was separated on a 1.5% agarose gel and bands were observed with the CL7, CL9 and CLl -specific primers but no products were observed for either the CL6 or the CLIO primers.
  • the CL7, CL9 and CLl products were purified using the QIAgen QIAquick PCR purification kit and cloned into pGEM-T-Easy. Positive clones were obtained and examined by DNA sequence analysis.
  • RNA was extracted from C. lugubris abdomen tissue using the method described above.
  • a cDNA library was constructed using the Invitrogen Cloneminer kit according to the manufacturer's instructions.
  • a fraction of the library was transformed into E. coli DHlOB ElectroMax competent cells (Invitrogen) and transformants selected on LB agar plates containing 25 ⁇ g/ml kanamycin.
  • Approximately 90% of the clones contained inserts with an average insert size of 1.5 kb.
  • Desaturase clones were identified and isolated from this library by PCR methods or by hybridisation screening using probes derived from the clones described above.
  • a volume of plasmid cDNA library representing approximately 50 colony forming units was seeded into 96 well plates containing 300 ⁇ l LB supplemented with 50 ⁇ g/ml kanamycin and grown overnight at 37 0 C with shaking.
  • lOO ⁇ l of each liquid culture from the wells across the plate were pooled into 1.5ml tubes to yield eight "lane pools” and a plasmid extraction was performed using a QIAquick miniprep spin kit (Qiagen) according to manufacturers instructions.
  • lO ⁇ l of each "lane pool” elutant was mixed into a 1.5ml tube creating a "plate pool".
  • PCR screening was done on each "plate pool” using gene specific primers and run out on 1% TAE agarose gel to identify positive plate pool/s. Once positive plate/s were identified PCR screening was repeated on each "lane pool” corresponding to a positive “plate pool” and run out on 1% TAE agarose gel to identify positive "lane pools", and in subsequent screening rounds on single wells and individual colonies to isolate positive clones. Plasmids which contained the gene of interest were analysed by restriction enzyme digest and sequence analysis.
  • Biotin labelled DNA probes were prepared using desaturase gene fragments labelled using dNTPs plus Biotin-dATP and DNA polymerase I Klenow fragment according to NEBlot phototope kit (New England Biolabs). Probes were hybridised to the membranes overnight at temperatures of 65 0 C (high stringency) or 55 0 C (moderate stringency). The hybridised biotinylated DNA was detected according to the Phototope-Star detection Kit manual (New England Biolabs) following the modification for colony hybridisations detailed in Appendix C. The membrane was exposed to Hyperf ⁇ lm ECL (Amersham Biosciences) for 1 minute and processed manually according to Amersham Biosciences recommendations. Positively hybridising clones were analysed by restriction enzyme digest and sequence analysis.
  • Example 8 Yeast expression vector construction and functional analysis of genes
  • Each of the full-length desaturase genes was expressed in Saccharomyces cerevisiae for functional characterization using the yeast expression vectors pYES2 or pVT100-U (Vernet, T et al. 1987). Each of the genomic regions containing putative desaturase genes was examined for restriction nuclease recognition sites. Restriction enzymes were chosen that did not cut the genomic regions but would facilitate directional cloning into pYES2 or pVT100-U. Primers were designed (Table 8) to the most 5' and 3' regions of the predicted desaturase genes with restriction sites at the 5' end of each of the primers to allow directional cloning.
  • DNAs of the yeast vectors were prepared and digested with the corresponding restriction enzymes and then dephosphorylated using Calf Intestinal Alkaline Phosphatase (Roche). After dephosphorylation, the digested vectors were purified using the QIAquick PCR purification kit (QIAgen). Gel extracted desaturase- containing fragments were ligated with digested vector and ligations transformed into E. coli DHlOB with transformants selected on LB agar plates with 100 ⁇ g/ml ampicillin. Clones containing inserts were confirmed by restriction enzyme analysis.
  • S. cerevisiae host strains Two S. cerevisiae host strains were used as recipients for pYES-derived constructs. These were S288C (genotype MAT ⁇ , SUC2 gal2 mal mel flol flo8-l hapl (Mortimer and Johnston (1986)) and OLEl (his3 ⁇ l, leu2 ⁇ 0, ura3 ⁇ 0, YMR272c::kanMX4) which is mutant for the gene encoding ⁇ 9-desaturase and could be used for complementation analysis. Five S. cerevisiae host strains were used as recipients for ⁇ VT-100 derived constructs.
  • each S. cerevisiae strain was streaked onto YPD (20g/l peptone, lOg/1 yeast extract, 2% glucose) and grown for several days at 3O 0 C. Transformations were performed using the Sigma Yeast Transformation kit according to manufacturer's instructions. Transformants were selected on SCMM-U agar plates (6.7 g/1 yeast nitrogen base without amino acids, 1.92 g/1 yeast synthetic drop-out media supplement without uracil, 2% glucose and 2% agar) at 30 0 C for up to 5 days. A number of transformants were selected for each construct and tested for the presence of plasmid DNA.
  • Control plates containing cis-10-heptadecenoic acid (1 mM) and 1% tergitol were also used to ensure yeast viability.
  • the results for the OLEl complementation tests for Tribolium and Chaulioganthus desaturases were as follows. Positive complementation was observed for Tribdesat2a, Tribdesat5, Tribdesat ⁇ a and Tribdesatl l, the last one with a lag phase. Possibly weak complementation was observed for Tribdesat2b and Tribdesat ⁇ . No complementation was observed for Tribdesatl, Tribdesat2c, Tribdesat4, TribdesatlO, Tribdesatl2, CLl and CL3.
  • Tribdesat2a, Tribdesat5, Tribdesat ⁇ a and Tribdesatl 1 were desaturases able to desaturate the native lipids of OLEl yeast cells to produce one of the complementing fatty acids and therefore were likely to be ⁇ 9 desaturases active on C16:0 or C18:0, or ⁇ ll desaturases active on C18:0, whereas Tribdesatl, Tribdesat2c, Tribdesat4, TribdesatlO, Tribdesatl2, CLl and CL3 were unlikely to encode one of these activities and therefore likely to encode a different desaturase.
  • Yeast transformants derived from stains S288C or OLEl were inoculated into 25 ml of synthetic minimal defined medium for yeast without Uracil (SCMM-U medium) containing 2% glucose and additionally 0.5 mM cis-11-heptadecenoic acid for the OLEl strain.
  • This inoculation culture was grown for 24 - 48 hr at 30°C with shaking. The OD600 of the culture was determined. From this, the amount of culture necessary to obtain an OD600 of 0.4 in 50ml of induction medium was calculated. This amount of inoculation culture was removed and the cells pelleted at 1500 x g for 5 minutes at 4 0 C.
  • the cells were then resuspended in 10 ml of induction medium, SCMM-U containing 2% galactose, 1% raffmose and 0.5 mM fatty acid substrate (added in ethanol/20% tergitol) and additionally 0.5 mM cis-11-heptadecenoic acid for OLEl derived transformants.
  • Each culture was grown for 24 - 48 h at 3O 0 C with shaking. Either 2 ml or the total culture was then used for analysis of fatty acids after harvesting cells by centrifugation, storage at -20 0 C if needed. Extraction of lipids from transformed S. cerevisiae and analysis
  • yeast cells from fatty acid feeding experiments were washed in 1% tergitol in water and pelleted at 1500 x g for 5 minutes at 4°C. Cell pellets (200-500mg) were then resuspended in water, transferred to glass test tubes and the cells again pelleted. Water was removed from the cell pellets in a Savant SpeedVac Plus SCIlOA concentrator/dryer. Each pellet was used either used directly or lipids were extracted with solvent as described below.
  • Lipids were extracted based on a Modified Folch Method (Protocol 7, pp22-24, Lipid Analysis, Hamilton and Hamilton, 1992). Lipids were extracted with 2 ml chloroform/methanol (2:1), and 0.5 ml of a saline solution, 0.9% w/v NaCl in water, was added. This was mixed thoroughly by vortexing. The layers were then allowed to separate; when a large amount of cell debris was present the samples were centrifuged at 2500 x g for 5 minutes. The top aqueous layer was removed and discarded. The bottom solvent layer was removed to a clean glass test tube. This was then dried down under nitrogen at 30°C.
  • Lipids were then derivatised prior to analysis by two different methylation methods. The first was the basic methylation of incorporated fatty acids, i.e. those found in triacyl glycerides or phospholipids, not free fatty acids, based on the method of Christie (2003).
  • the lipid sample was first dissolved in 0.5 ml hexane. Methyl acetate, 60 ⁇ l, was added, followed by 50 ⁇ l sodium methoxide at 0.5 M in methanol. The sealed test tube was then mixed thoroughly and placed in a heating block at 50°C for 10 minutes. After being allowed to cool for 5 minutes, a drop of acetic acid was added. The sample was dried in a gentle stream of nitrogen at 30°C, dissolved in 200 ⁇ l hexane and transferred to a vial prior to GC analysis.
  • the second method was an acidic methylation of all free and incorporated fatty acids, based on the method of Lewis et al. (2000).
  • a methanolic solution 2 ml of methanol/hydrochloric acid/ chloroform (10:1:1), was added to the dried yeast pellet or extracted lipid sample.
  • the sealed test tube was mixed thoroughly and placed in a heating block at 90°C for 60 minutes. After being allowed to cool for 10 minutes, 1 ml of 0.9% saline (NaCl in water) was added.
  • the methylated fatty acids were then extracted with 0.3 ml hexane. The hexane layer was transferred to a vial prior to analysis.
  • FAME samples were analysed by GC and GCMS.
  • GC analysis was carried out with a Varian 3800 gas chromatograph fitted with a flame ionisation detector (FID) and a BPX70 capillary column (length 30m, i.d. 0.32mm, film thickness 0.25 ⁇ m). Injections were made in the split mode using helium as the carrier gas and an initial column temperature of 100°C. The temperature was raised at 3°C/minute until 150°C, then raised at 5°C/minute until 170°C, held for 5 minutes, then raised at 50°C/minute until 255 0 C.
  • FAME fatty acid methyl ester
  • GC/MS analysis was carried out under similar chromatography conditions but with an initial column temperature of 60°C, raised at 20°C/minute until 170°C, held for 5 minutes, then raised at 50°C/minute until 255 0 C.
  • Detection was carried out using either a TEC PolarisQ Ion Trap or a Varian 1200 Single Quadrupole mass spectrometer. Mass spectra were acquired under positive electron impact in full scan mode between 50 - 400 aniu at the rate of 2 scans per sec. The mass spectra corresponding to each peal- in the chromatogram was automatically compared with spectra in the computerised NIST library.
  • Tribdesat2a had ⁇ 9 desaturase activity on saturated fatty acid carbon chain length from C10:0 to C16:0 when these fatty acids were fed to the yeast transformants. Activity was greatest on 14:0, but 15:0 and 16:0 were also efficient substrates. It had no detectable desaturase activity on 18:0, 20:0 or even longer saturated fatty acids in the yeast cells.
  • the protein had a predicted size of 320 amino acids and included the 8 conserved desaturase motifs and three His boxes (Table 3). On the basis of the sequence homology with known acyl-CoA desaturases, this enzyme was presumed to be acting on the acyl-CoA substrate. This enzyme was therefore characterised as a myristoyl-CoA ⁇ 9 desaturase.
  • Tnbdesatlb and Tr ⁇ bdesatlc were closely related proteins, having 64% amino acid sequence identity based on global alignment using BLOSUM62, which catalysed production of 5-hexadecenoic and 5-octadecenoic acids from palmitic acid and stearic acid, respectively. They were therefore characterised as ⁇ 5 desaturases acting on the saturated substrates C16:0 and C18:0.
  • the efficiency of conversion in yeast cells of C18:0 to C18:1 A5 was more than twice as efficient as the conversion of C16:0 to C16:1 ⁇ 5 (Table 9).
  • the enzymes were able to convert at least 8.0% of the substrate to the product in each case, to at least 40% for Tribdesat2b on C 18:0.
  • the enzymes did not detectably desaturate substrates of C14 or shorter when fed to the yeast cells, including C14:0 or C14:1 ⁇ 9 , or the substrates of C20 or longer including C20:0 or C20:l ⁇ 13 . Furthermore, the enzymes did not desaturate monounsaturated or polyunsaturated fatty acids including C16:1 ⁇ 9, C18:1 ⁇ 9, C18:2 or C18:3.
  • the proteins included the 8 conserved desaturase motifs and three His boxes (Table 3) and showed sequence homology with known acyl-CoA desaturases. They were therefore characterised as stearoyl-CoA ⁇ 5 desaturases. They also have palmitoyl-CoA ⁇ 5 desaturase activity.
  • AL21 was more active on the unsaturated substrate than the saturated substrate so it would not be considered as a stearoyl-CoA ⁇ 5 desaturase as defined herein.
  • the degree of identity in amino acid sequences when the proteins were compared over the entire sequences were: between AL21 and Tribdesat2b, 25%; AL21 and Tribdesat2b, 27%; ALlO and Tribdesat2b, 24%; ALlO and Trib2c, 28%. Therefore, it was concluded that there was insignificant or little homology between the plant acyl-CoA dependent desaturases and the insect ones.
  • Tribdesat ⁇ and 6a showed activity in yeast cells as ⁇ 9 desaturases that acted primarily and very efficiently on stearic acid to produce oleic acid. They also had some activity on palmitic acid to produce palmitoleic acid.
  • the enzymes did not detectably desaturate substrates of C 14 or shorter when fed to the yeast cells, including C14:0 or C14:1 ⁇ 9 , or the substrates of C20 or longer including C20:0. They were therefore characterised as stearoyl-CoA ⁇ 9 desaturases.
  • Tribdesat8 was able to desaturate palmitic acid to produce palmitoleic acid, so it was considered to be a ⁇ 9 desaturase. It did not have detectable desaturase activity in the yeast cells on C 14 or shorter substrates, or on C 18:0 or longer saturated substrates, or on monounsaturated or polyunsaturated substrates including C16:1 ⁇ 9 , C18:l , C18:2 or C18:3. It therefore appeared to be specific for palmitic acid.
  • the protein had a predicted size of 374 amino acids and included the 8 conserved desaturase motifs and three His boxes (Table 3). On the basis of the sequence homology with known acyl-CoA desaturases, this enzyme was presumed to be acting on the acyl-CoA substrate. This enzyme was therefore characterised as a palmitoyl- CoA ⁇ 9 desaturase.
  • TribdesatlO showed ⁇ 12 desaturase activity on palmitoleic and oleic acids resulting in C16:2 ⁇ 9> ⁇ 12 and C18:2 ⁇ 9> ⁇ 12 (LA), respectively.
  • Conversion of oleic acid substrate (at least 30% converted) was about twice as efficient in yeast cells compared to conversion of palmitoleic acid. There was no detectable activity on Cl 6:0, C 18:0 or longer saturated substrates, or Cl 4 or shorter substrates including C14:1 ⁇ 9 , so the enzyme appeared to be specific as a ⁇ 12 desaturase acting on ⁇ 9-mono-unsaturated substrates of Cl 8 and Cl 6 length joined to CoA.
  • the protein had a predicted size of 366 amino acids and included the 8 conserved desaturase motifs and three His boxes (Table 3).
  • this enzyme was presumed to be acting on acyl-CoA substrates.
  • This enzyme was therefore characterised as an oleoyl-CoA ⁇ 12 desaturase. It also showed palmitoleoyl- CoA ⁇ 12 desaturase activity. It was believed this is the first gene to be characterised encoding such an enzyme.
  • Tribdesatll showed ⁇ 9 desaturase activity on saturated substrates having a chain length from C14:0 to C24:0, with the greatest efficiency of conversion observed for C22:0 (known as behenic or docosanoic acid) and C24:0 (known as lignoceric or teracosanoic acid) . Therefore, this enzyme was considered to be a lignoceroyl-CoA ⁇ 9 desaturase, although it also has behenoyl-CoA ⁇ 9 desaturase activity.
  • CL3 showed efficient desaturation activity on C 14:0 to produce C14:1 ⁇ 9 .
  • the enzyme did not have detectable activity for C12:0 or C 16:0 and therefore appeared to be a specific myristoyl-CoA ⁇ 9 desaturase. It did not have activity on mono- unsaturated substrates such as C12:1 ⁇ 5 or C20:l.
  • this enzyme was presumed to be acting on the acyl-CoA substrate. This enzyme was therefore characterised as a myristoyl- CoA ⁇ 9 desaturase.
  • the pGem-T-Easy constructs containing full-length gene sequences were digested with restriction enzymes to excise the inserts. These were separated on a 1% agarose gel and the DNA of the approximately lkb fragments purified using the QIAquick Gel Extraction kit (Qiagen) according to the manufacturer's instructions.
  • the Gateway entry vector pENTR 11 (Invitrogen) was prepared and digested with the required restriction enzymes and dephosphorylated using Calf Intestine Alkaline Phosphatase (Roche). Gene fragments were ligated with the vector and transformed into E.coli strain JM109. Transformants were selected on LB agar plates containing 50 ⁇ g/ml kanamycin. Clones containing inserts were confirmed by restriction enzyme and sequence analysis.
  • Gateway expression vectors chosen for each.
  • pDEST8 was chosen and selection for DHlOBac transformants carried out in LB supplemented with kanamycin (50 ⁇ g/ml), gentamicin (7 ⁇ g/ml) and tetracycline (lO ⁇ g/ml), Bluo-gal (lOO ⁇ g/ml) and IPTG (40 ⁇ g/ml).
  • binary vector pXZP391 CSIRO Plant Industry
  • pENTRl 1 clones containing the gene inserts and appropriate pDEST vectors were prepared and under-went an LR recombination reaction according to manufacturers instructions (Gateway LR clonase enzyme mix, Invitrogen). The resultant recombination mixtures were transformed into E.coli strain JM 109 for pXZP391 or DHlOBac (Invitrogen) for pDEST ⁇ . Transformants containing correct inserts were confirmed by restriction enzyme analysis and sequence analysis using vector specific primers.
  • Example 10 Recombinant Baculovirus production of insect desaturases or acetylenases.
  • Tribdesat2b and CL3 genes were cloned into the pDest ⁇ vector
  • Transformants containing pFastbac plasmids having the 2b and CL2 inserts were grown overnight in LB medium containing the antibiotics.
  • a modified alkaline lysis plasmid isolation protocol was used according to the manufacturer's instructions in the Bac-to-Bac Expression System manual. Isolated DNA was dissolved in 40 ⁇ L of IX TE buffer pH 8.0 and quantitated using the Nanodrop spectrophotometer and stored at 4 0 C.
  • Sf9 i ⁇ podopetera frugiperda ovary cell line Sf9 i ⁇ podopetera frugiperda ovary cell line cells were seeded in a 6 well tissue culture plate at a rate of 9X10 5 cells per well, in 2mL of SF900II serum free medium (Invitrogen). The cells were allowed to adhere to the well surface for 2 hours at 27 0 C.
  • l ⁇ g of purified bacmid DNA was diluted in lOOuL of unsupplemented Grace's Medium (Invitrogen), in polystyrene tubes. 6 ⁇ L of Cellfectin® Reagent (Invitrogen) was diluted in lOO ⁇ L of unsupplemented Grace's Medium in polystyrene tubes.
  • the DNA and Cellfectin® Reagent were then mixed together and incubated at RT for 30 minutes. While DNA:lipid complexes were incubating, the cells were washed once with 2mL of unsupplemented Grace's Medium and the medium was then removed. 0.8mL of unsupplemented Grace's Medium was added to the DNA.iipid complex, mixed and then added to the washed SF9 cells. The transfection was allowed to proceed at 27 0 C for 5 hours. The DNA:lipid complexes were removed from the cells and 2mL of SF900II were added. Cells were incubated at 27 0 C until signs of infection were obvious. Once the cells appeared infected, the virus was harvested by centrifuging the cell culture medium at lKrpm for 5 minutes at 2O 0 C. The clarified cell culture medium was transferred to tubes and stored at 4 0 C. This was called Pl viral stock.
  • Sf9 cells were used to seed 25cm 2 flasks at a rate of lX10°cells/mL in 5mL of medium.
  • the cells were infected with Pl viral stock at a multiplicity of infection (MOI) of 0.1 virus particles per cell, which was estimated to be 5X10 6 cells/mL.
  • MOI multiplicity of infection
  • the P2 stock was then used to produce a P3 stock.
  • 75cm 2 flasks were seeded at a rate of lX10 6 cells/mL in 15 mL SF900II medium, and infected at a MOI of 0.1 virus particles per cell, which was estimated to be 5X10 6 cells/mL. After 48 hours at 27 0 C, the cells were showing signs of infection and the cell culture medium was harvested as outlined above. This was called P3 viral stock.
  • TCID 50 tissue Culture Infectious dose
  • a 96 well tray was seeded with 3X10 3 cells per well. Cells were incubated at 27 0 C overnight. 10-fold dilutions of the virus were prepared to 10 "8 . 50 ⁇ L of dilutions 10 "3 - 10 "8 were added to 8 wells each.
  • the TCID 50 was then incubated for 7 days at 27 0 C. The wells were scored positive or negative for virus infection. Once the viruses were titrated, infections of SF9 cells with known multiplicity of infections (MOI) could proceed. Infested cells were removed and dried under vacuum.
  • MOI multiplicity of infections
  • Methyl esters of fatty acids of insect cells infected with the construct expressing Tribdesat2b were obtained using the direct acidic methanol reagent as previously described and analysed by GC/MS.
  • the GC/MS trace and spectrum showed C16: 1 ⁇ 5 and C18:1 ⁇ 5 production by SF9 expressing Tribdesat2b, as compared to the SF9 control.
  • the efficiency of conversion of C16:0 to C16: ld5 in the insect cells transduced with the construct was 16.9% and the efficiency of conversion of C18:0 to C18:ld5 was 29.7%.
  • Tribolium TribdesatlO and 2b genes were cloned into pENTRl l as described above and then recombined into plant expression vector pXZP391 which provides for expression of the coding region insert under the control of a seed-specific napin promoter (FpI) isolated from Brassica napus (Stalberg et al., 1993).
  • FpI seed-specific napin promoter isolated from Brassica napus
  • Tl transformed plants were identified on MS media containing 40mg/L kanamycin and 100mg/L timentin, and transferred into pots in the glasshouse.
  • T2 seeds from each Tl plant were analysed for fatty acid composition by gas chromatography.
  • the data shown in Figure 6 demonstrated the ⁇ 12 desaturase activity of TribdesatlO in Arabidopsis, as LA was produced from oleic acid in the fad2/fael mutant plant.
  • Tribdesat 2b, CLl and CL2 coding regions were cloned in the 'sense' orientation into the plant expression vector pVEC8 (Wang et al., 1997) under the control of the regulatory sequences CaMV 35S promoter and octopine synthase transcription termination/polyadenylation signal (Gleave, 1992), providing strong constitutive expression of the transgene in most plant tissues.
  • the transgene was introduced into tobacco by Agrobacterium-mediaXed transformation of leaf tissue (Horsch et al., 1985) with selection of the transgenic cell lines on hygromycin- containing media.
  • Transgenic plant cells were selected based on the resistance to hygromycin, as conferred by a hpt resistance marker gene in the pVEC8 derived vector.
  • Transformed tobacco lines were cultured to produce either calli or regenerated to produce whole plants, depending on the phytohormone regime in the selection and growth media (Murashige and Skoog, 1962; Horsch et al., 1985).
  • Undifferentiated transgenic tobacco calli were produced by selection on calli-inducing phytohormones (0.5 mg/L indole acetic acid (IAA) and 0.05 mg/L kinetin) and calli were maintained on agar plates.
  • IAA indole acetic acid
  • Differentiated tobacco plants were produced by first selecting transgenic material on shoot-inducing media by the addition of shoot-inducing phytohormones (100 ⁇ g/L N-6-benzyladinine (BAP) and 500 ng/L L IAA). After 3 weeks of growth on this media individual tobacco apices were cut from the leaf discs and re-plated onto root-inducing media containing root-inducing phytohormones (50 ng/L IAA). A further 3 weeks are required for the formation of roots. Transgenic material, either calli or whole plants, were selected for analysis as required.
  • shoot-inducing phytohormones 100 ⁇ g/L N-6-benzyladinine (BAP) and 500 ng/L L IAA.
  • Tribdesat2b gene was also recombined using LR clonase into plant expression vector pXZP393 (CSIRO Plant Industry) under control of the constitutive CaMV 35S promoter.
  • the resulting expression plasmid pXZP384 was transformed into AGLl, and used for plant transformation into tobacco leaves as described above. Fatty acid composition of transgenic tobacco leaves and seed will be analysed by GC and GC-MS to shown modification of fatty acid composition by addition of the new desaturase activity.
  • Example 12 Cloning of desaturase genes from Acheta domesticus.
  • insects are thought to possess ⁇ 12-desaturase activity but as yet no gene encoding such an enzyme has been isolated despite extensive effort over 20 years. The reasons for this failure were unknown until the present work.
  • One insect known to possess ⁇ 12-desaturase activity is Acheta domesticus, the common cricket. In an initial attempt to clone an A.
  • AdD12Des-Fl (5'-TTGTTCTGTGTGGGTCAYGAYTGYGGWCA (SEQ ID NO: 127)) and AdD12Des-Rl (5'-GTGATGGGCGACGTGACYGTYKGTRAT (SEQ ID NO: 128) were designed based on the conserved histidine Box I and histidine Box III regions of A.
  • thaliana ⁇ 12-desaturase FAD2 P46313
  • Caenorhabditis elegans ⁇ 12- desaturase FAT-2 AAF63745
  • ⁇ 15-desaturase FAT-I L41807
  • RT-PCR reactions using A. domesticus fat body total RNA with this set of primers failed to amplify any specific product despite repeated attempts and varied conditions, suggesting that the A. domesticus gene(s) encoding ⁇ 12-desaturase might share low homology to the plant or nematode ⁇ 12-desaturase genes.
  • a different approach was considered to clone the A. domesticus ⁇ 12-desaturase gene. Based on the amino acid sequence homology shared among 55 insect fatty acid acyl-CoA ⁇ 9-, ⁇ 10-, ⁇ l l- and ⁇ 14-desaturases from species including Argyrotaenia velutinana (AAF44709), Bombyx mori (BAD18122, AAF80355), Epiphyas postvittana (AAK94070), Helicoverpa assult (AAM28484, AAM28483), H.
  • zea (AAF81787, AAF81788), Musca domestica (AAN31393), Ostrinia furnacalis (AAL27034, AAL32060, AAL35746), O. nubilalis (AAF44710, AAL35330, AAL35331), Trichoplusia ni (AAB92583, 044390, AAD03775) etc, degenerate primers (AdD12Des-F2 5'- TTCTTCTTCKCNCAYKTHGGNTGG (SEQ ID NO: 129) and AdD12Des-R2 5 '-TGRTGGTAGTTGTGVHANCCCTC (SEQ ID NO: 130)) were designed that targeted two conserved regions of these desaturases (FFFS/ AHI/VGW (SEQ ID NO: 131) and EGY/W/FHNYHH (SEQ ID NO: 132), corresponding to amino acid residues 145-152 and 263-270 of A.
  • velutinana ⁇ 9-desaturase AAF44709 in an attempt to clone an insect ⁇ 12-desaturase gene.
  • the cDNA sequences of AdD9Des, AdD12Des and TribdesatlO were each cloned in yeast expression vector pYES2 (Invitrogen), generating plasmids pXZP277, pXZP282 and pYES2-TriblO respectively. Plasmids pYES2, pXZP277, pXZP282 and pYES2-TriblO were transformed into Saccharomyces cerevisiae cell of the strain olel which contains a ⁇ 9-desaturase knock-out mutation, (Stukey et al., 1990) in order to test for complementation of this mutation and therefore indicate ⁇ 9-desaturase activity.
  • Yeast olel cells needed to be supplied with an unsaturated fatty acid such as C 16:1 ⁇ 9 to maintain growth, unless a ⁇ 9-desaturase is expressed within the cells, which was the basis of the complementation test.
  • Transformants in olel cells were selected on dropout media (SD-Ura) agar plate supplied with 0.5 mM C16:1 ⁇ 9 and 1% NP-40.
  • Yeast olel transformants carrying the above mentioned plasmids were first grown in YPD media (1% yeast extract, 2% peptone, 2% dextrose), supplied with 0.5 mM C16:1 ⁇ 9 and 1% NP-40 at 3O 0 C until OD 600 around 1.0.
  • T. castaneum ⁇ 12-desaturase When expressing in cells of yeast strain olel, T. castaneum ⁇ 12-desaturase produced C18:2 from C18:l at conversion rate of 30.5%, and C16:2 from C16:l at the conversion rate of 16.0% (Table 11), indicating a preference of the enzyme for the Cl 8 substrate over the C16 substrate. There was a concomitant decrease in the level of substrate fatty acid in the cells.
  • the A. domes ticus and T. castaneum ⁇ 12-desaturase protein coding regions were also cloned into a plant expression vector to generate plasmid pXZP375 and pXZP376 respectively, under control of the seed specific promoter FpI (Stalberg et al.,
  • AdDl 2Des gene encoding the A. domesticus ⁇ 12- desaturase efficiently produced C 18:2, with levels obtained of at least 42% of the total fatty acid in the seedoil, and at least 20% C18:3, both of which were produced in the seed from Cl 8:
  • the C18:3 produced in these transgenic lines was also a product of the conversion of C 18:1 to C 18:2 by the A. domes ticus ⁇ 12-desaturase, and represented A. domes ticus ⁇ 12-desaturase product.
  • This result confirmed the ⁇ 12- desaturase activity of the cloned gene, and represented the first demonstration of a recombinant insect ⁇ 12-desaturase in transformed cells.
  • castaneum TribdesatlO (SEQ ID NO: 28); TnD9, Trichoplusia ni AAB92583; TnDI l, T. ni 044390; other animal desaturases AcD12-15, Acanthamoeba castellanii ABK15557; CeD12, Caenorhabditis elegans AAF63745; RnD9, Rattus norvegicus P07308; bacterial desaturases N36D9, Nostoc sp. 36 CAF18423 ⁇ NoD9, Nostoc sp. PCC 7120 (Anabaena variabilis) BAA03434; NoD9, Nostoc sp.
  • D9, DlO, DI l, D12, and D14 designate ⁇ 9-, ⁇ 10-, ⁇ 11-, ⁇ 12-, and ⁇ 14- desaturases, respectively.
  • AdD12Des and TribdesatlO showed only low identities (up to 23%) over the full-length amino acid sequences to bacterial ⁇ 9-desarurases or plant, fungal and animal ⁇ 12-desaturases which formed another major cluster of predominantly acyl- lipid desaturases (left hand side of Figure 4).
  • the low level of identity of AdD12Des and TribdesatlO with ⁇ 12-desaturases from other animals, plants or fungi was particularly unexpected. Instead, AdD12Des formed a discrete cluster with insect ⁇ 9- desaturases including those of house cricket and red flour beetle suggesting its possible evolution from ⁇ 9-desaturase genes.
  • AdD12Des The extent of identity in amino acid sequence of AdD12Des to these were 65.2%, 55.2%, 55.5%, 53.5%, 53.5%, 55.3%, 59.4% and 53.6%, respectively, to insect ⁇ 9-desaturases from A. domesticus AF338466, D. melanogaster CAB69054, M. domesticus AAN31393, O. nubilalis AAF44710, P. octo AAF73073, T. castaneum XP_969607, T. castaneum XP_966962 and T. ni AAB9258.
  • AdD12Des was relatively close in sequence to the ⁇ 9-desaturase protein from the same species, sharing about 65% amino acid identity.
  • TribdesatlO appeared on a separate branch of the phylogenic tree to the other acyl CoA desaturases including Tribolium ⁇ 9-desaturases to which it shared up to 48% identity, it may have evolved independently of the insect ⁇ 9-desaturase genes.
  • AdD12Des retained low levels of ⁇ 9-desaturation activity on the medium chain saturated fatty acids, C 14:0 and Cl 5:0, when expressed in yeast.
  • Figure 3A shows the partial GC graph of fatty acid profile in yeast cells expressing pYES2 vector only, while 3B shows fatty acid profile in yeast cells expressing pXZP282, with extra C 14:1 and C 15:1 peaks.
  • acyl-CoA Cl 8:1 As substrate rather than acyl-PC Cl 8:1, the ⁇ 12-desaturase gene from A. domesticus in pXZP282 was expressed in yeast S288C cells.
  • the Arabidopsis thaliana ⁇ 12- desaturase gene coding region encoding FAD2 was also cloned into pYES2, resulted in the construct pXZP279, and expressed in S288C as a control gene, as an example of a plant ⁇ 12-desaturase that was known to be an acyl-PC type enzyme.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Nutrition Science (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Breeding Of Plants And Reproduction By Means Of Culturing (AREA)

Abstract

The present invention relates to enzymes which possess desaturase, conjugase, epoxidase and/or hydroxylase activity that can be used in methods of synthesizing fatty acids.

Description

SYNTHESIS OF FATTY ACIDS
FIELD OF THE INVENTION
The present invention relates to enzymes which possess desaturase, conjugase, epoxidase, and/or hydroxylase activity that can be used in methods of synthesizing fatty acids.
BACKGROUND OF THE INVENTION
The primary products of fatty acid biosynthesis in most organisms are 16- and 18-carbon compounds. However, the relative ratio of chain lengths and degree of unsaturation of these fatty acids vary widely among species. Mammals and insects, for example, produce primarily saturated and monosaturated fatty acids, while most higher plants produce fatty acids with one, two, or three double bonds, the latter two comprising polyunsaturated fatty acids (PUF A's).
Two main families of PUFAs are the omega-3 fatty acids (also represented as "n-3" fatty acids), exemplified by eicosapentaenoic acid (EPA, 20:5, n-3), and the omega-6 fatty acids (also represented as "n-6" fatty acids), exemplified by arachidonic acid (ARA, 20:4, n-6). PUFAs are important components of the plasma membranes of cells, predominantly esterified in the form of phospholipids, and adipose tissue in triglycerides.
The ability of cells to modulate the degree of unsaturation in their membranes is mainly determined by the action of fatty acid desaturases. Desaturase enzymes introduce unsaturated bonds at specific positions in their fatty acyl chain substrates. Desaturase enzymes generally show considerable selectivity both for the chain length of the substrate and for the location of existing double bonds in the fatty acyl chain (Shanklin and Cahoon, 1998) and may be classified on this basis. Another classification of fatty acid desaturases is based on the moiety to which the hydrocarbon chains of their substrates are acylated. Desaturases recognize substrates that are bound either to acyl carrier protein, to coenzyme A, or to lipid molecules such as phospholipids (Murata and Wada, 1995; Shanklin and Cahoon, 1998).
The desaturation of fatty acids in glycerolipids is essential for the proper function of biological membranes. Introduction of unsaturation in the Δ9 position of palmitic or stearic acid provides fluidity to membrane lipids and thus Δ9 desaturases are found universally in living systems.
Linoleic acid (LA; 18:2, Δ9, 12) is produced from oleic acid (18:1, Δ9) by a Δ12-desarurase while α-linolenic acid (ALA; 18:3) is produced from LA acid by a Δ15-desaturase. Stearidonic acid (18:4, Δ6, 9, 12, 15) and γ-linolenic acid (18:3, Δ6, 9, 12) are produced from ALA and LA, respectively, by a Δ6-desaturase. However, mammals cannot desaturate beyond the Δ9 position and therefore cannot convert oleic acid into LA. Fourteen insect species (de Renobales, 1987) have been shown to produce linoleic acid de novo from 14C-acetate, suggesting the presence of native Δ12-desaturase activity. The house cricket Acheta domesticus Δ12-desaturase activity was the first of this type reported to utilise oleoyl-CoA (Cripps, 1990). However, no gene responsible for the conversion of oleic acid to LA has been identified from an insect despite extensive effort. Likewise, ALA cannot be synthesized by mammals. The major poly-unsaturated fatty acids of animals therefore are derived from diet via the subsequent desaturation and elongation of dietary LA and ALA. Other eukaryotes, including fungi, nematodes and plants, have enzymes which desaturate at the carbon 12 and carbon 15 positions. The membrane-associated Δ12 desaturases of Arahidopsis sp. and soybean use acyl lipid substrates
Less commonly, saturated fatty acids can be unsaturated initially at positions other than the 9-position by desaturases with unusual specificities. Petroselinic acid (cis-6-octadecenoic acid) is concentrated in the seed oils of the Umbelliferae (Apiaceae), Araliaceae and Garryaceae plant families, where it can reach 85% of the total lipid fatty acid (Kleiman and Spencer, 1982). The desaturase from coriander (Coriandrum sativum) has been characterised that makes the unusual lipid. A plastid- located acyl-acyl carrier protein (ACP) Δ4 desaturase acts on ACP -palmitic acid to produce cis-4-hexadecenoic acid which is transferred from the plastid to the developing seed where it is elongated to cis-6-octadecenoic acid (Cahoon et al. 1992). A related desaturase with 83% sequence identity has been obtained from English Ivy (Hedra helix L.) which produces cis-4-hexadecenoic acid and cis-6-octadecenoic acid when expressed in Arabidopsis (Whittle et al 2005). cis-5- Eicosenoic acid (C20:l Δ5) is a major component of the seed oil of meadowfoam (Limnanthes alba) and related Limnathes species. The enzyme responsible for the production of the unusual oil in L. douglasii was identified as an acyl coenzyme A-Δ5 desaturase whose substrate preference is eicosanoic acid (Cahoon et al., 2000). Expression of L. douglasii Δ5 desaturase and fatty acid elongase genes in soybean embryos resulted in the production of cw-5-eicosenoic acid (C20:l Δ5) and cis-5 -docosenoic acid (C22:1Δ5). Sayanova et al (2007) have identified an acyl CoA-Δ5 desaturase which is related to the Limnathes sp. desaturase but it utilizes saturated fatty acids (C16:0, C 18:0) and unsaturated fatty acids (LA, ALA) to make Δ5 monoenoic acids and polyunsaturated fatty acids. Genes encoding similar enzymes have not been cloned from animals such as insects.
Some Lepidopteran insects carry out Δl l desaturation of saturated fatty acids (C16:O, C18:0) esterified to acylCoA as a step in the production of a diversity of moth sex pheromones (Rodriguez et al 2004). Desaturases from the moth Spodoptera littoralis were expressed in Saccharomyces cerevisiae to produce cis-Δl l mono- unsaturated products of C14:0, C16:0 and C18:0 when the yeast cells were fed additional saturated fatty acids (Rodriguez et al 2004). In addition, trans-Δl l tetradecenoic acid was formed from myristic acid (C 14:0) fed to the yeast. A minor byproduct of the Δl l desaturation was the formation of 11 -hydroxy hexadecanoic or octadecanoic acid (up to 0.1% of total fatty acids) (Serra et al., 2006). Moto et al. (2004) identified a bi-functional acyl-CoA desaturase from the pheromone gland of the silkmoth which was responsible for the biosynthesis of the pheromone precursor. The desaturase first utilised palmitic acid to make cis-11-hexadecenoic acid and then acted on this to remove allylic 2H and form a conjugated diene fatty acid (trans- Δ10,cis-Δ12-hexadecendienoic acid and some trans-Δ10,trans-Δ12-hexadecendienoic acid) and therefore it possessed both cis-Δl l and conjugase desaturase activities, hi the New Zealand leaf roller, Plαnotortrix octo, a desaturase has been identified from pheromone gland that desaturates palmitic acid at the Δ10 position to form cis-ΔlO- hexadecenoic acid (Hao et al. 2002).
Omega-3 LC-PUFA are now widely recognized as important compounds for human and animal health and the inclusion of omega-3 LC-PUFA such as EPA and DHA in the human diet has been linked with numerous health-related benefits. These include prevention or reduction of coronary heart disease, hypertension, type-2 diabetes, renal disease, rheumatoid arthritis, ulcerative colitis, chronic obstructive pulmonary disease, various mental disorders such as schizophrenia, attention deficit hyperactive disorder and Alzheimer's disease, and aiding brain development and growth (Simopoulos, 2000). These fatty acids may be obtained from dietary sources or by conversion of linoleic (LA, omega-6) or α-linolenic (ALA, omega-3) fatty acids, both of which are regarded as essential fatty acids in the human diet. While humans and many other vertebrate animals are able to convert LA or ALA, obtained from plant sources, to LC-PUFA, they carry out this conversion at a very low rate. Moreover, most modern societies have unbalanced diets in which at least 90% of polyunsaturated fatty acid(s) consist of omega-6 fatty acids, instead of the 4:1 ratio or less for omega-6: omega-3 fatty acids that is regarded as ideal (Trautwein, 2001). The immediate dietary source of LC-PUFA such as eicosapentaenoic acid (EPA, 20:5) and docosahexaenoic acid (DHA5 22:6) for humans is mostly from fish or fish oil. Health professionals have therefore recommended the regular inclusion of fish containing significant levels of LC-PUFA into the human diet. Increasingly, fish-derived LC- PUFA oils are being incorporated into food products and in infant formula. However, due to a decline in global and national fisheries, alternative sources of these beneficial health-enhancing oils are needed. Fatty acids may also be hydroxylated, for example ricinoleic acid (12-hydroxy- octadec-cw-9-enoic acid) which comprises up to 90% of the fatty acid in castor oil from Ricinus communis and is an important agricultural commodity oil. Other related hydroylated fatty acids found in plant oils include 12-hydroxy-octadeca-c«-9,cz,s'~15- dienoic (densipolic) and 14-hydroxy-eicosa-cw-ll,cw-17-dienoic (auricolic) acids. The Ricinus Δ12 hydroxylase acts on oleic acid lipid substrate to produce ricinoleic acid; the desaturase gene responsible for the transformation is most closely related to but divergent from plant membrane Δ12 acyl lipid desaturases (van de Loo et al 1995). There is a homologous C20 hydroxylated fatty acid produced at high levels in seed oil of Lesquerella sp. as 14-hydroxy-eicos-cw-l l-enoic (lesquerolic acid) (Gunstone et al., 1994). The L.fendleri hydroxylase gene has been cloned and expressed in an Arabidopsis FAD2 mutant which accumulated ricinoleic, lesquerolic and densipolic acids in seeds (Broun et al 1998). 2-hydroxy fatty acids occur in appreciable amounts in the sphingolipids of plants and animals but they are also present as minor components of seeds oils such as 2-hydroxy-octadeca-9,12,15- trienoate from Thymus vulgaris seeds and 2-hydroxy-oleic and linoleic acids are found in Salvia nilotica (Smith, 1971; Badami and Patil, 1981).
Fatty acids may also comprise epoxy groups. The most widely known natural epoxy fatty acid is vernolic acid (12,13-epoxy-octadec-cw-9-enoic acid) from the seed oils of Vernonia spp and Euphorbia lagascae (Cuperus and Derksen, 1996). The epoxygenase gene of C. palaestina, which is related to but divergent from plant membrane Δ12-oleate desaturases, has been functionally characterized and shown to use linoleate as a substrate (Lee et al. 1998).
In some organisms, conjugated fatty acids are produced by the activity of a conjugase (Crombie et al., 1984; Crombie et al., 1985; Fritsche et al., 1999; Cahoon et al., 2001; Qiu et al., 2001). The biosynthesis of conjugated fatty acids such as calendulic acid, eleostearic acid or punicic acid proceeds via the desaturation of oleic acid to linoleic acid by a Δ12-desaturase and a further desaturation in conjunction with a rearrangement of the Z9- or Z12-double bond to the conjutrienic fatty acid by a specific conjutriene-forming desaturase (conjugase).
There is a need for further methods of producing fatty acids in recombinant cells and for more efficient production or production of novel fatty acids.
Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context for the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed before the priority date of each claim of this application. SUMMARY OF THE INVENTION
The present inventors have identified novel enzymes having desaturase, conjugase, epoxygenase, and/or hydroxylase activity from insects of the Order Coleoptera and Orthoptera.
Accordinlgy, the present invention provides a eukaryotic cell comprising an exogenous nucleic acid encoding a polypeptide which is:
(i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs :16 to 30, 73 to 78, 80 and 134,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and/or 134, and/or
(iii) a biologically active fragment of i) or ii), wherein the polypeptide has one or more activities selected from desaturase, conjugase, epoxidase and hydroxylase activity.
In one embodiment, the polypeptide can be isolated from an insect of the Order Coleoptera or Orthoptera. Examples of insect species from which the polypeptide can be isolated include, but are not limited to, Tribolium, Chauliognathus or Acheta.
In one embodiment, the polypeptide encoded by the exogenous nucleic acid is:
(i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs: 28, 73, and/or 134,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134, and/or
(iii) a biologically active fragment of (i) or (ii), wherein the polypeptide has acyl-CoA Δ12 desaturase activity.
In another embodiment, the polypeptide is:
(i) a polypeptide comprising amino acids having a sequence as set forth in SEQ ID NO: 18 or SEQ ID NO: 19,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to a sequence set forth in SEQ ID NO: 18 or SEQ ID NO: 19, and/or
(iii) a biologically active fragment of (i) or (ii), wherein the polypeptide has acyl-CoA Δ5 desaturase activity.
In yet another embodiment, the polypeptide is:
(i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs: 17, 22, 23, 27, 29, 74, 75 and/or 76, (ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 17, 22, 23, 27, 29, 74, 75 and/or 76, and/or
(iii) a biologically active fragment of (i) or (ii), wherein the polypeptide has acyl-CoA Δ9 desaturase activity.
The present inventors are the first to identify a nucleic acid encoding an an acyl-CoA Δ12 desaturase. Thus, the present invention provides a eukaryotic cell comprising an exogenous nucleic acid encoding an acyl-CoA Δ12 desaturase.
In one embodiment, the acyl-CoA Δ12 desaturase comprises:
(i) amino acids having a sequence as set forth in any one of SEQ ID NOs: 28, 73, and/or 134,
(ii) amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134, and/or
(iii) a biologically active fragment of (i) or (ii).
In one embodiment, the eukaryotic cell comprises an increased level of 16:2Δ9'Δ12 and/or 18:2Δ9'A12 fatty acids relative to a corresponding eukaryotic cell lacking the exogenous nucleic acid.
In yet another embodiment, the eukaryotic cell comprises an increased level of 16:2Δ9'Δ12 and/orl8:2Δ9'Δ12 fatty acids which are esterified to CoA.
The present inventors are also the first to identify an acyl-CoA Δ5 desaturase which has a preference for a 18:0 and/or 16:0 fatty acid substrate when compared to a number of other substrates. Accordingly, in a further aspect the present invention provides a eukaryotic cell comprising an exogenous nucleic acid encoding an acyl- CoA Δ5 desaturase, wherein the desaturase is more active on a 18:0 and/or 16:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is any one, two, three or all of a 18:1Δ9, 16:1A9, 20:0 and 20:2Δ11Δ14.
In one embodiment, the acyl-CoA Δ5 desaturase comprises:
(i) amino acids having a sequence as set forth in SEQ ID NO: 18 or SEQ ID NO: 19,
(ii) amino acids having a sequence which is at least 50% identical to SEQ ID NO: 18 or SEQ ID NO: 19, and/or
(iii) a biologically active fragment of (i) or (ii).
In another embodiment, the eukaryotic cell comprises an increased level of 16:1 5 and/or 18:1A5 fatty acids relative to a corresponding eukaryotic cell lacking the exogenous nucleic acid.
In yet another embodiment, the eukaryotic cell comprises an increased level of 16:1Δ5 and/orl8:lΔ5 fatty acids which are esterified to CoA. The present inventors are also the first to identify an acyl-CoA Δ9 desaturase which is more active on a 14:0 substrate than on certain fatty acid substrates esterified to CoA. Thus, in a further aspect the present invention provides a eukaryotic cell comprising an exogenous nucleic acid encoding an acyl-CoA Δ9 desaturase, wherein the desaturase is more active on a 14:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is 16:0 and/or 18:0.
In an embodiment, the acyl-CoA Δ9 desaturase comprises:
(i) amino acids having a sequence as set forth in SEQ ID NO: 23 or SEQ ID NO: 74,
(ii) amino acids having a sequence which is at least 50% identical to SEQ ID NO: 23 or SEQ ID NO:74, and/or
(iii) a biologically active fragment of (i) or (ii).
In one particular embodiment, the eukaryotic cell comprises an increased level of 14:1 A9 relative to a corresponding eukaryotic cell lacking the exogenous nucleic acid.
In yet another embodiment, the eukaryotic cell comprises an increased level of 14:1Δ9 which is esterified to CoA.
Preferably, the eukaryotic cell comprising the exogenous nucleic acid is a plant cell, a mammalian cell, an insect cell, a fungal cell or a yeast cell.
In one embodiment, the eukaryotic cell is in a plant or plant seed.
Preferably, the plant or plant seed is an oilseed plant or an oilseed respectively.
In one aspect of the invention there is provided a process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
(i) obtaining a nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence more closely related to SEQ ID NO: 28 than to SEQ ID NO: 135,
(ii) introducing the nucleic acid molecule into a cell or cell-free expression system in which the promoter is active,
(iii) determining whether the fatty acid composition is modified relative to the cell or cell-free expression system before introduction of the nucleic acid molecule, and
(iv) optionally, selecting a nucleic acid molecule which modified the fatty acid composition.
In another aspect the present invention provides a process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
(i) obtaining a nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence that is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134,
(ii) introducing the nucleic acid molecule into a cell or cell-free expression system in which the promoter is active,
(iii) determining whether the fatty acid composition is modified relative to the cell or cell-free expression system before introduction of the nucleic acid molecule, and
(iv) optionally, selecting a nucleic acid molecule which modified the fatty acid composition.
In one embodiment, step (iv) comprises selecting a nucleic acid molecule encoding an acyl-CoA Δ12 desaturase.
In another embodiment, the modified fatty acid composition comprises an increased level of 16:2Δ9'Δ12 and/or 18 :2Δ9Λ12.
The present invention further provides a process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
(i) obtaining a nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence that is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 18 and/or 19,
(ii) introducing the nucleic acid molecule into a cell or cell-free expression system in which the promoter is active,
(iii) determining whether the fatty acid composition is modified relative to the cell or cell-free expression system before introduction of the nucleic acid, and
(iv) optionally, selecting a nucleic acid molecule which modified the fatty acid composition.
In one embodiment, step (iv) comprises selecting a nucleic acid molecule an acyl-CoA Δ5 desaturase, wherein the desaturase is more active on a 18:0 and/or 16:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is any one, two, three or all of a 18:1Δ9, 16:1Δ9, 20:0 and 20:2Δ11Δ14.
In another embodiment, the modified fatty acid composition comprises an increased level of 16:1Δ5 and/or 18:1Δ5 fatty acids.
The present invention further provides a process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
(i) obtaining a nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence that is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 17, 22, 23, 27, 29, 74, 75 and/or 76, (ii) introducing the nucleic acid molecule into a cell or cell-free expression system in which the promoter is active,
(iii) determining whether the fatty acid composition is modified relative to the cell or cell-free expression system before introduction of the nucleic acid, and
(iv) optionally, selecting a nucleic acid molecule which modified the fatty acid composition.
In one embodiment, step (iv) comprises selecting a nucleic acid molecule encoding an acyl-CoA Δ9 desaturase, wherein the desaturase is more active on a 14:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is 16:0 and/or 18:0.
In another embodiment, the modified fatty acid composition comprises an increased level of 14: 1Δ9.
In yet another embodiment, the polypeptide encoded by the nucleic acid molecule is an insect polypeptide or mutant thereof.
The present invention further provides a substantially purified or recombinant polypeptide which is an acyl-CoA Δ12 desaturase.
In one embodiment, the polypeptide is:
(i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs: 28, 73, and/or 134,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134, and/or
(iii) a biologically active fragment of (i) or (ii).
The present invention further provides a substantially purified or recombinant polypeptide which is an acyl-CoA Δ5 desaturase, wherein the desaturase is more active on a 18:0 and/or 16:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is any one, two, three or all of a 18: 1Δ9, 16:1 Δ9, 20:0 and
Figure imgf000010_0001
In one embodiment, the polypeptide is:
(i) a polypeptide comprising amino acids having a sequence as set forth in SEQ ID NO: 18 or SEQ ID NO:195
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to SEQ ID NO: 18 and/or SEQ ID NO: 19, and/or
(iii) a biologically active fragment of (i) or (ii).
The present invention further provides a substantially purified or recombinant polypeptide which is an acyl-CoA Δ9 desaturase, wherein the desaturase is more active on a 14:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is 16:0 and/or 18:0. In one embodiment, the polypeptide is:
(i) a polypeptide comprising amino acids having a sequence as set forth in SEQ ID NO: 17 or SEQ ID NO:74,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to one or more of the sequences as set forth in SEQ ID NO: 17 and/or SEQ ID NO: 74, and/or
(iii) a biologically active fragment of (i) or (ii).
The present invention further provides a substantially purified or recombinant polypeptide which is:
(i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs :16 to 30, 73 to 78, 80 and 134,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and/or 134, and/or
(iii) a biologically active fragment of i) or ii), wherein the polypeptide has one or more an activities selected from desaturase, conjugase, epoxidase and/or hydroxylase activity.
Preferably, the polypeptide comprises amino acids having a sequence which is at least 90% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and/or 134.
In one embodiment, the polypeptide can be isolated from an insect of the Order Coleoptera or Orthoptera.
Preferably, the polypeptide has desaturase activity upon a carbon-carbon bond located at any one of the Δ2 to Δ15 positions of a fatty acid.
In one embodiment, the polypeptide has desaturase activity on a C16 or C18 fatty acid.
In another embodiment, the polypeptide is a fusion protein further comprising at least one other polypeptide sequence.
The present invention further provides an isolated and/or exogenous polynucleotide comprising:
(i) a sequence of nucleotides selected from any one of SEQ ID NOs: 1 to 15, 67 to 72, 79 and 133,
(ii) a sequence of nucleotides encoding a polypeptide of the invention,
(iiϊ) a sequence of nucleotides which are at least 50% identical to one or more of the sequences set forth in SEQ ID NOs: 1 to 15, 67 to 72, 79 and/or 133, and/or
(iv) a sequence which hybridises to any one of (i) to (iii) under stringent conditions.
Also provided is a vector comprising a polynucleotide of the invention. In one embodiment, the polynucleotide is operably linked to a promoter.
The present invention further provides a cell comprising the recombinant polypeptide according to the invention, the exogenous polynucleotide of the invention and/or the vector of the invention.
Preferably, the cell is a plant, fungal, yeast, bacterial or animal cell. More preferably, the cell is a eukaryote cell.
The present invention further provides a method of producing the polypeptide according to the invention, the method comprising expressing in a cell or cell free expression system the polynucleotide of the invention.
In one embodiment, the method further comprises isolating the polypeptide.
The present invention further provides a transgenic non-human organism comprising a cell according to the invention.
In one embodiment, the organism is a transgenic plant.
The present invention further provides a seed comprising the cell according to the invention.
The present invention further provides oil produced by, or obtained from, the cell according to the invention, the transgenic non-human organism of the invention, or the seed of the invention.
In one embodiment, the oil comprises fatty acids 16:0, 16:1A5, 18:0 and 18:1Λ5, wherein the ratio of the total amount of 18: 1Δ5 to 18:0 in the oil is between 100:1 and 1:2, and wherein the fatty acid of the oil comprises less than 10%, or less than 5% (w/w) 20:lΔ5.
In another embodiment, at least 43%, or at leasst 50%, or at least 60%, of the C18 fatty acid of the oil is 18 :1Δ5.
In yet another embodiment, the fatty acid of the oil comprises at least 3.0% (w/w), or at least 5% (w/w) or at least 10% (w/w), 18 : 1 Δ5 as a percentage of the total fatty acid of the oil.
In another embodiment, the oil comprises less than 10% (w/w), or less than 5% (w/w), 18:3Δ9Λ12'Δ15 (ALA) as a percentage of the total fatty acid in the oil.
In one embodiment, the oil is obtained by extraction of oil from an oilseed.
The present invention further provides an oil comprising fatty acids 16:0, 16:1Δ5, 18:0 and 18:1Δ5, wherein the ratio of the total amount of 18:1Δ5 to 18:0 in the oil is between 100:1 and 1:2, and wherein the fatty acid of the oil comprises less than 10% (w/w), or less than 5%, 20: 1A5.
The present invention further provides an oil comprising fatty acids, wherein at least 43%, or at leasst 50%, or at least 60%, of the C18 fatty acid of the oil is 18: ;11 ΔS The invention further provides an oil comprising fatty acids comprising at least 3.0% (w/w), or at least 5% (w/w) or at least 10% (w/w), 18:1Δ5 as a percentage of the total fatty acid of the oil.
In one embodiment, oil comprises less than 10% (w/w), or less than 5% (w/w), 18:3Δ9'Δ12'Δ15 (ALA) as a percentage of the total fatty acid in the oil.
The present invention further provides a fatty acid produced by, or obtained from, the cell according to the invention, the transgenic non-human organism of the invention, or the seed of the invention.
The invention also provides a method of producing oil containing unsaturated fatty acids, the method comprising extracting oil from the cell according to the invention, the transgenic non-human organism of the invention, or the seed of the invention.
The present invention further provides a composition comprising a cell according to the invention, the polypeptide according to the invention, a polynucleotide according to the invention, a vector of the invention, an oil according to the invention or a fatty acid of the invention.
The invention also provides feedstuffs, cosmetics or chemicals comprising the oil according to the invention or a fatty acid of the invention.
The present invention also provides a method of performing a desaturase reaction, the method comprising contacting a substrate saturated, monounsaturated or polyunsaturated fatty acid esterified to CoA with the polypeptide of the invention
The present invention futher provides a substantially purified antibody, or fragment thereof, that specifically binds a polypeptide of the invention
The invention further provides a method of treating or preventing a condition which would benefit from a PUFA, the method comprising administering to a subject a cell according to the invention, the polypeptide according to the invention, a polynucleotide according to the invention, a vector of the invention, an oil according to the invention, a fatty acid of the invention and/or a feedstuff of the invention.
In one embodiment, the condition is cardiac arrhythmia's, angioplasty, inflammation, asthma, psoriasis, osteoporosis, kidney stones, AIDS, multiple sclerosis, rheumatoid arthritis, Crohn's disease, schizophrenia, cancer, foetal alcohol syndrome, attention deficient hyperactivity disorder, cystic fibrosis, phenylketonuria, unipolar depression, aggressive hostility, adrenoleukodystophy, coronary heart disease, hypertension, diabetes, obesity, Alzheimer's disease, chronic obstructive pulmonary disease, ulcerative colitis, restenosis after angioplasty, eczema, high blood pressure, platelet aggregation, gastrointestinal bleeding, endometriosis, premenstrual syndrome, myalgic encephalomyelitis, chronic fatigue after viral infections or an ocular disease. The present invention also provides use of a cell according to the invention, the polypeptide according to the invention, a polynucleotide according to the invention, a vector of the invention, an oil according the invention or a fatty acid of the invention and/or a feedstuff of the invention for the manufacture of a medicament for treating or preventing a condition which would benefit from a PUFA.
As will be apparent, preferred features and characteristics of one aspect of the invention are applicable to many other aspects of the invention.
Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
The invention is hereinafter described by way of the following non-limiting Examples and with reference to the accompanying figures.
BRIEF DESCRIPTION OF THE ACCOMPANYING DRAWINGS
Figure 1. A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 2b. Dots indicate sequence identity, while different nucleotides are indicated.
Figure 2. A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 3. Dots indicate sequence identity, while different nucleotides are indicated.
Figure 3. A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 6b. Dots indicate sequence identity, while different nucleotides are indicated.
Figure 4. A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 10. Dots indicate sequence identity, while different nucleotides are indicated.
Figure 5. A comparison of the nucleic acid sequence obtained from the gene prediction program and that of the authentic RT-PCR product for Tribdesat 11. Dots indicate sequence identity, while different nucleotides are indicated.
Figure 6. GC/MS trace and spectrum showing linoleic acid (18:2 Δ9,Δ12) production by Tribdesat 10 in Arabidopsis, with Arabidopsis Fadl/Fae2 control.
Figure 7. A, Gas chromatography (GC) of yeast fatty acid methyl esters from S. cerivisiae expressing AdDl 2Des. B. Gas chromatography (GC) of yeast fatty acid methyl esters from S. cerivisiae, vector only. C. Confirmation of the double bond positions of C16:2 and C18:2 products by GC-mass spectrometry. Figure 8. GC analysis of yeast olel cells expressing pYES2 vector only (Panel A) or A. domesticus Δ12-desaturase in pXZP282 (Panel B) after fed with mixture of C14:0 and C15:0.
Figure 9. Phylogenetic analysis of representative acyl-CoA or acyl-lipid desaturase protein sequences.
Figure 10. Phylogenetic analysis of representative acyl-CoA or acyl-lipid desaturase protein sequences.
KEY TO THE SEQUENCE LISTING
SEQ ID NO: 1 - coding sequence of Tribolium desaturase 1 SEQ ID NO: 2 - coding sequence of Tribolium desaturase 2a SEQ ID NO: 3 - coding sequence of Tribolium desaturase 2b SEQ ID NO: 4 - coding sequence of Tribolium desaturase 2c SEQ ID NO: 5 — coding sequence of Tribolium desaturase 3 SEQ ID NO: 6 - coding sequence of Tribolium desaturase 4 SEQ ID NO: 7 — coding sequence of Tribolium desaturase 5 SEQ ID NO: 8 — coding sequence of Tribolium desaturase 6a SEQ ID NO: 9 - coding sequence of Tribolium desaturase 6b SEQ ID NO: 10 - coding sequence of Tribolium desaturase 7a SEQ ID NO: 11 - coding sequence of Tribolium desaturase 7b SEQ ID NO: 12 - coding sequence of Tribolium desaturase 8 SEQ ID NO: 13 - coding sequence of Tribolium desaturase 10 SEQ ID NO: 14 - coding sequence of Tribolium desaturase 11 SEQ ID NO: 15 - coding sequence of Tribolium desaturase 12 SEQ ID NO: 16 - amino acid sequence of Tribolium desaturase 1 SEQ ID NO: 17 - amino acid sequence of Tribolium desaturase 2a SEQ ID NO: 18 - amino acid sequence of Tribolium desaturase 2b SEQ ID NO: 19 - amino acid sequence of Tribolium desaturase 2c SEQ ID NO: 20 - amino acid sequence of Tribolium desaturase 3 SEQ ID NO: 21 — amino acid sequence of Tribolium desaturase 4 SEQ ID NO: 22 - amino acid sequence of Tribolium desaturase 5 SEQ ID NO: 23 - amino acid sequence of Tribolium desaturase 6a SEQ ID NO: 24 - amino acid sequence of Tribolium desaturase 6b SEQ ID NO: 25 - amino acid sequence of Tribolium desaturase 7a SEQ ID NO: 26 - amino acid sequence of Tribolium desaturase 7b SEQ ID NO: 27 - amino acid sequence of Tribolium desaturase 8 SEQ ID NO: 28 - amino acid sequence of Tribolium desaturase 10 SEQ ID NO: 29 - amino acid sequence of Tribolium desaturase 11 SEQ ID NO: 30 — amino acid sequence of Tribolium desaturase 12 SEQ ID NO: 31 - conserved insect desaturase sequence SEQ ID NOs: 32 to 66 - oligonucleotide primers SEQ ID NO: 67 — coding sequence of Chauliognathus desaturase CLl SEQ ID NO: 68 - coding sequence of Chauliognathus desaturase CL3 SEQ ID NO: 69 - partial coding sequence of Chauliognathus desaturase CL6 SEQ ID NO: 70 -partial coding sequence of Chauliognathus desaturase CL7 SEQ ID NO: 71 - partial coding sequence of Chauliognathus desaturase CL 8 SEQ ID NO: 72 - partial coding sequence of Chauliognathus desaturase CL9 SEQ ID NO: 73 - amino acid sequence of Chauliognathus desaturase CLl SEQ ID NO: 74 - amino acid sequence of Chauliognathus desaturase CL3 SEQ ID NO: 75 - partial amino acid sequence of Chauliognathus desaturase CL6 SEQ ID NO: 76 — partial amino acid sequence of Chauliognathus desaturase CL7 SEQ ID NO: 77 -partial amino acid sequence of Chauliognathus desaturase CL8 SEQ ID NO: 78 — partial amino acid sequence of Chauliognathus desaturase CL9 SEQ ID NO: 79 — coding sequence of Chauliognathus desaturase CNl SEQ ID NO: 80 — amino acid sequence of Chauliognathus desaturase CNl SEQ ID NOs: 81 to 103 - oligonucleotide primers SEQ ID NOs: 104 to 111 - conserved desaturase motifs SEQ ID NOs: 112 to 122 - histidine boxes in desaturase sequences SEQ ID NOs: 123 to 126 - signature motifs SEQ ID NOs: 127 to 130 - oligonucleotides SEQ ID NOs: 131 and 132 - conserved regions
SEQ ID NO: 133 — coding sequence of Acheta domesticus desaturase AdDIIDes SEQ ID NO: 134 - amino acid sequence of Acheta domesticus desaturase AdDl 2Des SEQ ID NO: 135 -Arabidopsis thaliana FAD2 Δ12 desaturase
DETAILED DESCRIPTION OF THE INVENTION
General Techniques
Unless specifically defined otherwise, all technical and scientific terms used herein shall be taken to have the same meaning as commonly understood by one of ordinary skill in the art (e.g., in cell culture, molecular genetics, immunology, immunohistochemistry, protein chemistry, and biochemistry).
Unless otherwise indicated, the recombinant protein, cell culture, and immunological techniques utilized in the present invention are standard procedures, well known to those skilled in the art. Such techniques are described and explained throughout the literature in sources such as, J. Perbal, A Practical Guide to Molecular Cloning, John Wiley and Sons (1984), J. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbour Laboratory Press (1989), T. A. Brown (editor), Essential Molecular Biology: A Practical Approach, Volumes 1 and 2, IRL Press (1991), D.M. Glover and B.D. Hames (editors), DNA Cloning: A Practical Approach, Volumes 1-4, IRL Press (1995 and 1996), and F.M. Ausubel et al. (editors), Current Protocols in Molecular Biology, Greene Pub. Associates and Wiley- Interscience (1988, including all updates until present), Ed Harlow and David Lane (editors) Antibodies: A Laboratory Manual, Cold Spring Harbour Laboratory, (1988), and J.E. Coligan et al. (editors) Current Protocols in Immunology, John Wiley & Sons (including all updates until present), and are incorporated herein by reference.
Selected Definitions
As used herein, the term "fatty acid" refers to a carboxylic acid (or organic acid), often with a long aliphatic tail, either saturated or unsaturated. Typically fatty acids have a carbon-carbon bonded chain of at least 8 carbon atoms in length, more preferably at least 12 carbons in length. Most naturally occurring fatty acids have an even number of carbon atoms because their biosynthesis involves acetate which has two carbon atoms. The fatty acids may be in a free state (non-esterified) or in an esterified form such as part of a triglyceride, diacylglyceride, monoacylglyceride, acyl-CoA (thio-ester) bound or other bound form. The fatty acid may be esterified as a phospholipid such as a phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylglycerol, phosphatidylinositol or diphosphatidylglycerol forms.
"Saturated fatty acids" do not contain any double bonds or other functional groups along the chain. The term "saturated" refers to hydrogen, in that all carbons (apart from the carboxylic acid [-COOH] group) contain as many hydrogens as possible. In other words, the omega (ω) end contains 3 hydrogens (CH3-) and each carbon within the chain contains 2 hydrogens (-CH2-).
"Unsaturated fatty acids" are of similar form to saturated fatty acids, except that one or more alkene functional groups exist along the chain, with each alkene substituting a singly-bonded " -CH2-CH2-" part of the chain with a doubly-bonded "- CH=CH-" portion (that is, a carbon double bonded to another carbon). The two next carbon atoms in the chain that are bound to either side of the double bond can occur in a cis or trans configuration.
As used herein, the terms "monounsaturated fatty acid" refers to a fatty acid which comprises at least 12 carbon atoms in its carbon chain and only one alkene group in the chain. As used herein, the terms "polyunsaturated fatty acid" or "PUFA" refer to a fatty acid which comprises at least 12 carbon atoms in its carbon chain and at least two alkene groups (carbon-carbon double bonds). Ordinarily, the number of carbon atoms in the carbon chain of the fatty acids refers to an unbranched carbon chain. If the carbon chain is branched, the number of carbon atoms excludes those in sidegroups. In one embodiment, the long-chain polyunsaturated fatty acid is an ω3 fatty acid, that is, having a desaturation (carbon-carbon double bond) in the third carbon-carbon bond from the methyl end of the fatty acid. In another embodiment, the long-chain polyunsaturated fatty acid is an ω6 fatty acid, that is, having a desaturation (carbon-carbon double bond) in the sixth carbon-carbon bond from the methyl end of the fatty acid.
As used herein, the terms "long-chain polyunsaturated fatty acid" or "LC- PUFA" refer to a fatty acid which comprises at least 20 carbon atoms in its carbon chain and at least two carbon-carbon double bonds.
As used herein, the term "desaturase" refers to an enzyme which is capable of introducing a carbon-carbon double bond into the acyl group of a fatty acid substrate, which is typically in an esterified form such as, for example, fatty acid CoA esters. The acyl group may be esterified to a phospholipid such as phosphatidyl choline, or to acyl carrier protein (ACP), or in a preferred embodiment to CoA. Desaturases generally may be categorized into three groups accordingly.
As used herein, the term "Δ12 desaturase" refers to a protein which performs a desaturase reaction that introduces a carbon-carbon double bond located at the 12th bond from the carboxyl end and has greater activity in desaturation at this position than any other position, hi one embodiment, the Δ12 desaturase activity includes oleoyl-CoA Δ12 desaturase activity. In another embodiment, the Δ12 desaturase activity includes palmitoleoyl-CoA Δ12 desaturase activitiy. These fatty acids may be in an esterified form, such as, for example, as part of a phospholipid. Examples of Δ12 desaturases include proteins comprising an amino acid sequence provided in SEQ ID NOs: 28, 78 and 134.
There are two types of Δ12 desaturases; acyl-CoA Δ12 desaturases and acyl- PC Δ12 desaturases, which are predominantly active on acyl-CoA and acyl-PC linked 18:1 substrates, respectively. However, Δ9 desaturases may also be active on an acyl- ACP (acyl carrier protein).
As used herein, the term "acyl-CoA Δ12 desaturase activity" or "acyl-CoA Δ12 desaturase" refers to the desaturase having greater activity on an acyl-CoA substrate than an acyl-lipid (such as acyl-PC) and/or acyl-ACP substrate. In an embodiment, the activity is at least two-fold greater.
As used herein, a "Δ5 desaturase" refers to a protein which performs a desaturase reaction that introduces a carbon-carbon bond located at the 5th bond from the carboxyl end and has greater activity in desaturation at this position than any other position, hi one embodiment the Δ5 desaturase has acyl-CoA-stearoyl Δ5 desaturase activity. In another embodiment, the Δ5 desaturase has acyl-CoA-palmitoyl Δ5 desaturase activity. In one embodiment, the enzyme Δ5 desaturase catalyses the desaturation of C20 LC-PUFA, converting dihomo-γ-linoleic acid DGLA to arachidonic acid (ARA, 20:4ω6) and ETA to EPA (20:5ω3).
As used herein, the term "acyl-CoA Δ5 desaturase activity" or "acyl-CoA Δ5 desaturase" refers to the desaturase having greater activity on an acyl-CoA substrate than an acyl-lipid (such as acyl-PC) and/or acyl-ACP substrate. In an embodiment, the activity is at least two-fold greater.
As used herein, a "Δ9 desaturase" refers to a protein which performs a desaturase reaction that introduces a carbon-carbon bond located at the 9 bond from the carboxyl end and has greater activity in desaturation at this position than any other position. Examples of Δ9 desaturase activity include myristoyl-CoA Δ9 desaturase activity, stearoyl-CoA Δ9 desaturase activity, palmitoyl-CoA Δ9 desaturase activity, lignoceroyl-CoA Δ9 desaturase activity and behenoyl-CoA Δ9 desaturase activity.
As used herein, the term "acyl-CoA Δ9 desaturase activity" or "acyl-CoA Δ9 desaturase" refers to the desaturase having greater activity on an acyl-CoA substrate than an acyl-lipid (such as acyl-PC) and/or acyl-ACP substrate. In an embodiment, the activity is at least two-fold greater.
As used herein, the term "conjugase" refers to a coηjutriene-forming desaturase.
The term "epoxidase" as used herein refers to an enzyme that introduces an epoxy group into a fatty acid resulting in the production of an epoxy fatty acid. In preferred embodiment, the epoxy group is introduced at the 2nd and/or 12th carbon on a fatty acid chain, especially ofa C16 or C18 fatty acid chain.
"Hydroxylase", as used herein, refers to an enzyme that introduces a hydroxyl group into a fatty acid resulting in the production of a hydroxylated fatty acid. In a preferred embodiment, the hydroxyl group is introduced at the 2nd, 12th and/or 17th carbon on a Cl 8 fatty acid chain. In another preferred embodiment, the hydroxyl group is introduced at the 15th carbon on a Cl 6 fatty acid chain.
The term "plant" includes whole plants, vegetative structures (for example, leaves, stems), roots, floral organs/structures, seed (including embryo, endosperm, and seed coat), plant tissue (for example, vascular tissue, ground tissue, and the like), cells and progeny of the same.
A "transgenic plant", "genetically modified plant" or variations thereof refers to a plant that contains a gene construct ("transgene") not found in a wild-type plant of the same species, variety or cultivar. A "transgene" as referred to herein has the normal meaning in the art of biotechnology and includes a genetic sequence which has been produced or altered by recombinant DNA or RNA technology and which has been introduced into the plant cell. The transgene may include genetic sequences derived from a plant cell. Typically, the transgene has been introduced into the plant by human manipulation such as, for example, by transformation but any method can be used as one of skill in the art recognizes.
The terms "seed" and "grain" are used interchangeably herein. "Grain" generally refers to mature, harvested grain but can also refer to grain after imbibition or germination, according to the context. Mature grain commonly has a moisture content of less than about 18-20%.
"Operably linked" as used herein refers to a functional relationship between two or more nucleic acid (e.g., DNA) segments. Typically, it refers to the functional relationship of transcriptional regulatory element (promoter) to a transcribed sequence. For example, a promoter is operably linked to a coding sequence, such as a polynucleotide defined herein, if it stimulates or modulates the transcription of the coding sequence in an appropriate cell. Generally, promoter transcriptional regulatory elements that are operably linked to a transcribed sequence are physically contiguous to the transcribed sequence, i.e., they are cw-acting. However, some transcriptional regulatory elements, such as enhancers, need not be physically contiguous or located in close proximity to the coding sequences whose transcription they enhance.
As used herein, the term "gene" is to be taken in its broadest context and includes the deoxyribonucleotide sequences comprising the protein coding region of a structural gene and including sequences located adjacent to the coding region on both the 5' and 3' ends for a distance of at least about 2 kb on either end and which are involved in expression of the gene. The sequences which are located 5' of the coding region and which are present on the niRNA are referred to as 5' non-translated sequences. The sequences which are located 3' or downstream of the coding region and which are present on the rnRNA are referred to as 3' non-translated sequences. The term "gene" encompasses both cDNA and genomic forms of a gene. A genomic form or clone of a gene contains the coding region which may be interrupted with non-coding sequences termed "introns" or "intervening regions" or "intervening sequences." Introns are segments of a gene which are transcribed into nuclear RNA (hnRNA); introns may contain regulatory elements such as enhancers. Introns are removed or "spliced out" from the nuclear or primary transcript; introns therefore are absent in the messenger RNA (rnRNA) transcript. The mRNA functions during translation to specify the sequence or order of amino acids in a nascent polypeptide. The term "gene" includes a synthetic or fusion molecule encoding all or part of the proteins of the invention described herein and a complementary nucleotide sequence to any one of the above. As used herein, the term "can be isolated from" means that the polynucleotide or encoded polypeptide is naturally produced by an organism, particularly an insect.
Polypeptides/Peptides
By "substantially purified polypeptide" or "purified polypeptide" we mean a polypeptide that has generally been separated from the lipids, nucleic acids, other peptides, and other contaminating molecules with which it is associated in its native state. Preferably, the substantially purified polypeptide is at least 60% free, more preferably at least 75% free, and more preferably at least 90% free from other components with which it is naturally associated.
The term "recombinant" in the context of a polypeptide refers to the polypeptide when produced by a cell, or in a cell-free expression system, in an altered amount or at an altered rate compared to its native state. In one embodiment the cell is a cell that does not naturally produce the polypeptide. However, the cell may be a cell which comprises a non-endogenous gene that causes an altered amount of the polypeptide to be produced. A recombinant polypeptide of the invention includes polypeptides which have not been separated from other components of the transgenic (recombinant) cell, or cell-free expression system, in which it is produced, and polypeptides produced in such cells or cell-free systems which are subsequently purified away from at least some other components.
The terms "polypeptide" and "protein" are generally used interchangeably.
The % identity of a polypeptide is determined by GAP (Needleman and Wunsch, 1970) analysis (GCG program) with a gap creation penalty=5, and a gap extension penalty=0.3. The query sequence is at least 15 amino acids in length, and the GAP analysis aligns the two sequences over a region of at least 15 amino acids. More preferably, the query sequence is at least 50 amino acids in length, and the GAP analysis aligns the two sequences over a region of at least 50 amino acids. More preferably, the query sequence is at least 100 amino acids in length and the GAP analysis aligns the two sequences over a region of at least 100 amino acids. Even more preferably, the query sequence is at least 250 amino acids in length and the GAP analysis aligns the two sequences over a region of at least 250 amino acids. Even more preferably, the GAP analysis aligns two sequences over their entire length.
As used herein a "biologically active" fragment is a portion of a polypeptide of the invention which maintains a defined activity of the full-length polypeptide, namely possessing desaturase, conjugase, epoxidase, and/or hydroxylase activity. Biologically active fragments can be any size as long as they maintain the defined activity. Preferably, the biologically active fragment maintains at least 10% of the acivity of the full length protein. With regard to a defined polypeptide/enzyme, it will be appreciated that % identity figures higher than those provided above will encompass preferred embodiments. Thus, where applicable, in light of the minimum % identity figures, it is preferred that the polypeptide/enzyme comprises an amino acid sequence which is at least 60%, more preferably at least 65%, more preferably at least 70%, more preferably at least 75%, more preferably at least 76%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 91%, more preferably at least 92%, more preferably at least 93%, more preferably at least 94%, more preferably at least 95%, more preferably at least 96%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99%, more preferably at least 99.1%, more preferably at least 99.2%, more preferably at least 99.3%, more preferably at least 99.4%, more preferably at least 99.5%, more preferably at least 99.6%, more preferably at least 99.7%, more preferably at least 99.8%, and even more preferably at least 99.9% identical to the relevant nominated SEQ ID NO.
With regard to the acyl-CoA Δ5 desaturases of the invention, this is the first demonstration of an animal desaturase that can produce 5-hexadecenoic and 5- octadecenoic acids and uses an acyl-CoA fatty acid as a substrate.
With regard to the acyl-CoA Δ12 desaturases of the invention, this is the first known demonstration of of Δ12 desaturase activity by an enzyme naturally produced by an animal. Furthermore, this is the first demonstration of a desaturase that can produce linoleic acid and uses an acyl-CoA fatty acid as a substrate.
Amino acid sequence mutants of the polypeptides of the present invention can be prepared by introducing appropriate nucleotide changes into a nucleic acid of the present invention, or by in vitro synthesis of the desired polypeptide. Such mutants include, for example, deletions, insertions or substitutions of residues within the amino acid sequence. A combination of deletion, insertion and substitution can be made to arrive at the final construct, provided that the final peptide product possesses the desired characteristics.
Mutant (altered) peptides can be prepared using any technique known in the art. For example, a polynucleotide of the invention can be subjected to in vitro mutagenesis. Such in vitro mutagenesis techniques include sub-cloning the polynucleotide into a suitable vector, transforming the vector into a "mutator" strain such as the E. coli XL-I red (Stratagene) and propagating the transformed bacteria for a suitable number of generations. In another example, the polynucleotides of the invention are subjected to DNA shuffling techniques as broadly described by Harayama (1998). Products derived from mutated/altered DNA can readily be screened using techniques described herein to determine if they possess desaturase, conjugase, epoxidase, and/or hydroxylase activity.
In designing amino acid sequence mutants, the location of the mutation site and the nature of the mutation will depend on characteristic(s) to be modified. The sites for mutation can be modified individually or in series, e.g., by (1) substituting first with conservative amino acid choices and then with more radical selections depending upon the results achieved, (2) deleting the target residue, or (3) inserting other residues adjacent to the located site.
Amino acid sequence deletions generally range from about 1 to 15 residues, more preferably about 1 to 10 residues and typically about 1 to 5 contiguous residues.
Substitution mutants have at least one amino acid residue in the polypeptide molecule removed and a different residue inserted in its place. The sites of greatest interest for substitutional mutagenesis include sites identified as the active site(s). Other sites of interest are those in which particular residues obtained from various strains or species are identical. These positions may be important for biological activity. These sites, especially those falling within a sequence of at least three other identically conserved sites, are preferably substituted in a relatively conservative manner. Such conservative substitutions are shown in Table 1 under the heading of "exemplary substitutions".
Table 1 - Exemplary substitutions.
Figure imgf000023_0001
Figure imgf000024_0001
In a preferred embodiment a mutant/variant polypeptide has one or two or three or four conservative amino acid changes when compared to a naturally occurring polypeptide. Details of conservative amino acid changes are provided in Table 1. In a preferred embodiment, the changes are not in one or more of the following motifs: AGAHRLW, SETDAD, FFFSHVG, QKKY, NSAAH, GEGWHNYHH, PWDY5 and GWAY. In a particularly preferred embodiment, the changes are not in each of the following motifs: AGAHRLW, SETDAD, FFFSHVG, QKKY, NSAAH, GEGWHNYHH, PWDY, and GWAY. As the skilled person would be aware, such minor changes can reasonably be predicted not to alter the activity of the polypeptide when expressed in a recombinant cell.
Furthermore, if desired, unnatural amino acids or chemical amino acid analogues can be introduced as a substitution or addition into the polypeptides of the present invention. Such amino acids include, but are not limited to, the D-isomers of the common amino acids, 2,4-diaminobutyric acid, α-amino isobutyric acid, 4- aminobutyric acid, 2-aminobutyric acid, 6-amino hexanoic acid, 2-amino isobutyric acid, 3 -amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, β-alanine, fluoro-amino acids, designer amino acids such as β-methyl amino acids, Cα-methyl amino acids, Nα-methyl amino acids, and amino acid analogues in general.
Also included within the scope of the invention are polypeptides of the present invention which are differentially modified during or after synthesis, e.g., by biotinylation, benzylation, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. These modifications may serve to increase the stability and/or bioactivity of the polypeptide of the invention.
Polypeptides of the present invention can be produced in a variety of ways, including production and recovery of natural polypeptides, production and recovery of recombinant polypeptides, and chemical synthesis of the polypeptides. In one embodiment, an isolated polypeptide of the present invention is produced by culturing a cell capable of expressing the polypeptide under conditions effective to produce the polypeptide, and recovering the polypeptide. A preferred cell to culture is a recombinant cell of the present invention. Effective culture conditions include, but are not limited to, effective media, bioreactor, temperature, pH and oxygen conditions that permit polypeptide production. An effective medium refers to any medium in which a cell is cultured to produce a polypeptide of the present invention. Such medium typically comprises an aqueous medium having assimilable carbon, nitrogen and phosphate sources, and appropriate salts, minerals, metals and other nutrients, such as vitamins. Cells of the present invention can be cultured in conventional fermentation bioreactors, shake flasks, test tubes, microtiter dishes, and petri plates. Culturing can be carried out at a temperature, pH and oxygen content appropriate for a recombinant cell. Such culturing conditions are within the expertise of one of ordinary skill in the art.
Polynucleotides
By "isolated polynucleotide" we mean a polynucleotide which has generally been separated from the polynucleotide sequences with which it is associated or linked in its native state. Preferably, the isolated polynucleotide is at least 60% free, more preferably at least 75% free, and more preferably at least 90% free from other components with which it is naturally associated. Furthermore, the term "polynucleotide" is used interchangeably herein with the terms "nucleic acid molecule", "gene" and "mRNA".
The term "exogenous" in the context of a polynucleotide refers to the polynucleotide when present in a cell, or in a cell-free expression system, in an altered amount compared to its native state. In one embodiment, the cell is a cell that does not naturally comprise the polynucleotide. However, the cell may be a cell which comprises a non-endogenous polynucleotide resulting in an altered amount of production of the encoded polypeptide. An exogenous polynucleotide of the invention includes polynucleotides which have not been separated from other components of the transgenic (recombinant) cell, or cell-free expression system, in which it is present, and polynucleotides produced in such cells or cell-free systems which are subsequently purified away from at least some other components. The exogenous polynucleotide (nucleic acid) can be a contiguous stretch of nucleotides existing in nature, or comprise two or more contiguous stretches of nucleotides from different sources (naturally occurring and/or synthetic) joined to form a single polynucleotide. Typically such chimeric polynucleotides comprise at least an open reading frame encoding a polypeptide of the invention operably linked to a promoter suitable of driving transcription of the open reading frame in a cell of interest. "Polynucleotide" refers to a oligonucleotide, polynucleotide or any fragment thereof. It may be DNA or RNA of genomic or synthetic origin, double-stranded or single-stranded, and combined with carbohydrate, lipids, protein, or other materials to perform a particular activity defined herein.
The % identity of a polynucleotide is determined by GAP (Needleman and Wunsch, 1970) analysis (GCG program) with a gap creation penalty=5, and a gap extension penalty=0.3. The query sequence is at least 45 nucleotides in length, and the GAP analysis aligns the two sequences over a region of at least 45 nucleotides. Preferably, the query sequence is at least 150 nucleotides in length, and the GAP analysis aligns the two sequences over a region of at least 150 nucleotides. Even more preferably, the query sequence is at least 300 nucleotides in length and the GAP analysis aligns the two sequences over a region of at least 300 nucleotides. Even more preferably, the GAP analysis aligns two sequences over their entire length.
With regard to the defined polynucleotides, it will be appreciated that % identity figures higher than those provided above will encompass preferred embodiments. Thus, where applicable, in light of the minimum % identity figures, it is preferred that the polynucleotide comprises a polynucleotide sequence which is at least 60%, more preferably at least 65%, more preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 91%, more preferably at least 92%, more preferably at least 93%, more preferably at least 94%, more preferably at least 95%, more preferably at least 96%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99%, more preferably at least 99.1%, more preferably at least 99.2%, more preferably at least 99.3%, more preferably at least 99.4%, more preferably at least 99.5%, more preferably at least 99.6%, more preferably at least 99.7%, more preferably at least 99.8%, and even more preferably at least 99.9% identical to the relevant nominated SEQ ID NO.
A polynucleotide of the present invention may selectively hybridise to a polynucleotide that encodes a polypeptide of the present invention under stringent conditions. As used herein, under stringent conditions are those that (1) employ low ionic strength and high temperature for washing, for example, 0.015 M NaCl/0.0015 M sodium citrate/0.1% NaDodSO4 at 500C; (2) employ during hybridisation a denaturing agent such as formamide, for example, 50% (vol/vol) formamide with 0.1% bovine serum albumin, 0.1% Ficoll, 0.1% polyvinylpyrrolidone, 50 mM sodium phosphate buffer at pH 6.5 with 750 mM NaCl, 75 mM sodium citrate at 420C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 g/ml), 0.1% SDS and 10% dextran sulfate at 420C in 0.2 x SSC and 0.1% SDS.
Polynucleotides of the present invention may possess, when compared to naturally occurring molecules, one or more mutations which are deletions, insertions, or substitutions of nucleotide residues. Mutants can be either naturally occurring (that is to say, isolated from a natural source) or synthetic (for example, by performing site- directed mutagenesis or DNA shuffling on the nucleic acid as described above). It is thus apparent that polynucleotides of the invention can be either naturally occurring or recombinant.
Oligonucleotides of the present invention can be RNA, DNA, or derivatives of either. The minimum size of such oligonucleotides is the size required for the formation of a stable hybrid between an oligonucleotide and a complementary sequence on a polynucleotide of the present invention. Preferably, the oligonucleotides are at least 15 nucleotides, more preferably at least 18 nucleotides, more preferably at least 19 nucleotides, more preferably at least 20 nucleotides, even more preferably at least 25 nucleotides in length. The present invention includes oligonucleotides that can be used as, for example, probes to identify nucleic acid molecules, or primers to produce nucleic acid molecules. Oligonucleotides of the present invention used as a probe are typically conjugated with a label such as a radioisotope, an enzyme, biotin, a fluorescent molecule or a chemiluminescent molecule.
Recombinant Vectors
One embodiment of the present invention includes a recombinant vector, which comprises at least one isolated polynucleotide molecule of the present invention, inserted into any vector capable of delivering the polynucleotide molecule into a host cell. Such a vector contains heterologous polynucleotide sequences, that is polynucleotide sequences that are not naturally found adjacent to polynucleotide molecules of the present invention and that preferably are derived from a species other than the species from which the polynucleotide molecule(s) are derived. The vector can be either RNA or DNA, either prokaryotic or eukaryotic, and typically is a transposon (such as described in US 5,792,294), a virus or a plasmid.
One type of recombinant vector comprises a polynucleotide molecule of the present invention operatively linked to an expression vector. The phrase "operably linked" refers to insertion of a polynucleotide molecule into an expression vector in a manner such that the molecule is able to be expressed when transformed into a host cell. As used herein, an expression vector is a DNA or RNA vector that is capable of transforming a host cell and of effecting expression of a specified polynucleotide molecule. Preferably, the expression vector is also capable of replicating within the host cell. Expression vectors can be either prokaryotic or eukaryotic, and are typically viruses or plasmids. Expression vectors of the present invention include any vectors that function (i.e., direct gene expression) in recombinant cells of the present invention, including in bacterial, fungal, endoparasite, arthropod, animal, and plant cells. Particularly preferred expression vectors of the present invention can direct gene expression in yeast and/or plants cells.
In particular, expression vectors of the present invention contain regulatory sequences such as transcription control sequences, translation control sequences, origins of replication, and other regulatory sequences that are compatible with the recombinant cell and that control the expression of polynucleotide molecules of the present invention. In particular, recombinant molecules of the present invention include transcription control sequences. Transcription control sequences are sequences which control the initiation, elongation, and termination of transcription. Particularly important transcription control sequences are those which control transcription initiation, such as promoter, enhancer, operator and repressor sequences. Suitable transcription control sequences include any transcription control sequence that can function in at least one of the recombinant cells of the present invention. A variety of such transcription control sequences are known to those skilled in the art. Particularly preferred transcription control sequences are promoters active in directing transcription in plants, either constitutively or stage and/or tissue specific, depending on the use of the plant or parts thereof. These plant promoters include, but are not limited to, promoters showing constitutive expression, such as the 35S promoter of Cauliflower Mosaic Virus (CaMV), those for fruit-specific expression, such as the polygalacturonase (PG) promoter from tomato.
Recombinant molecules of the present invention may also (a) contain secretory signals (i.e., signal segment nucleic acid sequences) to enable an expressed polypeptide of the present invention to be secreted from the cell that produces the polypeptide and/or (b) contain fusion sequences which lead to the expression of nucleic acid molecules of the present invention as fusion proteins. Examples of suitable signal segments include any signal segment capable of directing the secretion of a polypeptide of the present invention. Preferred signal segments include, but are not limited to, Nicotiana nectarin signal peptide (US 5,939,288), tobacco extensin signal, the soy oleosin oil body binding protein signal. In addition, a nucleic acid molecule of the present invention can be joined to a fusion segment that directs the encoded polypeptide to the proteosome, such as a ubiquitin fusion segment. Recombinant molecules may also include intervening and/or untranslated sequences surrounding and/or within the nucleic acid sequences of nucleic acid molecules of the present invention.
Host Cells
Another embodiment of the present invention includes a recombinant cell comprising a host cell transformed with one or more recombinant molecules of the present invention. Transformation of a polynucleotide molecule into a cell can be accomplished by any method by which a polynucleotide molecule can be inserted into the cell. Transformation techniques include, but are not limited to, transfection, electroporation, microinjection, lipofection, adsorption, and protoplast fusion. A recombinant cell may remain unicellular or may grow into a tissue, organ or a multicellular organism. Transformed polynucleotide molecules of the present invention can remain extrachromosomal or can integrate into one or more sites within a chromosome of the transformed (i.e., recombinant) cell in such a manner that their ability to be expressed is retained.
Suitable host cells to transform include any cell that can be transformed with a polynucleotide of the present invention. Host cells of the present invention either can be endogenously (i.e., naturally) capable of producing polypeptides of the present invention or can be capable of producing such polypeptides after being transformed with at least one polynucleotide molecule of the present invention. Host cells of the present invention can be any cell capable of producing at least one protein of the present invention, and include plant, bacterial, fungal (including yeast), parasite, and arthropod cells. Preferably, the host cell is a plant, arthropod or yeast cell. Non limiting examples of arthropod cells include insect cells such as Spodoptera frugiperda (Sf) cells, e.g. Sf9, Sf21, Trichoplusia ni cells, and Drosophila S2 cells.
An example of a bacterial cell useful as a host cell of the present invention is Synechococcus spp. (also known as Synechocystis spp.), for example Synechococcus elongatus.
The cells may be of an organism suitable for a fermentation process. As used herein, the term the "fermentation process" refers to any fermentation process or any process comprising a fermentation step. A fermentation process includes, without limitation, fermentation processes used to produce alcohols (e.g., ethanol, methanol, butanol); organic acids (e.g., citric acid, acetic acid, itaconic acid, lactic acid, gluconic acid); ketones (e.g., acetone); amino acids (e.g., glutamic acid); gases (e.g., H2 and CO2); antibiotics (e.g., penicillin and tetracycline); enzymes; vitamins (e.g., riboflavin, beta-carotene); and hormones. Fermentation processes also include fermentation processes used in the consumable alcohol industry (e.g., beer and wine), dairy industry (e.g., fermented dairy products), leather industry and tobacco industry. Preferred fermentation processes include alcohol fermentation processes, as are well known in the art. Preferred fermentation processes are anaerobic fermentation processes, as are well known in the art.
Suitable fermenting cells, typically microorganisms are able to ferment, i.e., convert, sugars, such as glucose or maltose, directly or indirectly into the desired fermentation product. Examples of fermenting microorganisms include fungal organisms, such as yeast. As used herein, "yeast" includes Saccharomyces spp., Saccharomyces cerevisiae, Saccharomyces carlbergensis, Candida spp., Kluveromyces spp., Pichia spp., Hansenula spp., Trichoderma spp., Lipomyces starkey, and Yarrowia lipolytica. Preferred yeast include strains of the Saccharomyces spp., and in particular, Saccharomyces cerevisiae. Commercially available yeast include, e.g., Red Star/Lesaffre Ethanol Red (available from Red Star/Lesaffre, USA) FALI (available from Fleischmann's Yeast, a division of Burns Philp Food Inc., USA), SUPERSTART (available from Alltech), GERT STRAND (available from Gert Strand AB, Sweden) and FERMIOL (available from DSM Specialties).
Recombinant DNA technologies can be used to improve expression of a transformed polynucleotide molecule by manipulating, for example, the number of copies of the polynucleotide molecule within a host cell, the efficiency with which those polynucleotide molecules are transcribed, the efficiency with which the resultant transcripts are translated, and the efficiency of post-translational modifications. Recombinant techniques useful for increasing the expression of polynucleotide molecules of the present invention include, but are not limited to, operatively linking polynucleotide molecules to high-copy number plasmids, integration of the polynucleotide molecule into one or more host cell chromosomes, addition of vector stability sequences to plasmids, substitutions or modifications of transcription control signals (e.g., promoters, operators, enhancers), substitutions or modifications of translational control signals (e.g., ribosome binding sites, Shine-Dalgarno sequences), modification of polynucleotide molecules of the present invention to correspond to the codon usage of the host cell, and the deletion of sequences that destabilize transcripts.
Transgenic Plants
The term "plant" as used herein as a noun refers to whole plants, but as used as an adjective refers to any substance which is present in, obtained from, derived from, or related to a plant, such as for example, plant organs (e.g. leaves, stems, roots, flowers), single cells (e.g. pollen), seeds, plant cells and the like. Plants provided by or contemplated for use in the practice of the present invention include both monocotyledons and dicotyledons. In preferred embodiments, the plants of the present invention are crop plants (for example, cereals and pulses, maize, wheat, potatoes, tapioca, rice, sorghum, millet, cassava, barley, or pea), or other legumes. The plants may be grown for production of edible roots, tubers, leaves, stems, flowers or fruit. The plants may be vegetables or ornamental plants. The plants of the invention may be: corn (Zea mays), canola {Brassica napiis, Brassica rapa ssp.), flax (Linum usitatissimum), alfalfa (Medicago sativά), rice (Oryza sativa), rye (Secede cerale), sorghum {Sorghum bicolour, Sorghum vulgare), sunflower (Helianthus annus), wheat (Tritium aestivum), soybean (Glycine max), tobacco (Nicotiana tabacum), potato (Solarium tuberosum), peanuts (Arachis hypogaea), cotton (Gossypium hirsutum), sweet potato (Lopmoea batatus), cassava (Manihot esculenta), coffee (Cofea spp.), coconut (Cocos nucifera), pineapple (Anana comosus), citris tree (Citrus spp.), cocoa (Theobroma cacao), tea (Camellia senensis), banana (Musa spp.), avocado (Persea americana), fig (Ficus casica), guava (Psidium guajava), mango (Mangifer indica), olive (Olea europaea), papaya (Carica papaya), cashew (Anacardium occidentale), macadamia (Macadamia intergrifolia), almond (Prunus amygdalus), sugar beets (Beta vulgaris), oats, or barley.
In a preferred embodiment, the plant is an angiosperm.
In one embodiment, the plant is an oilseed plant, preferably an oilseed crop plant. As used herein, an "oilseed plant" is a plant species used for the commercial production of oils from the seeds of the plant. The oilseed plant may be oil-seed rape (such as canola), maize, sunflower, soybean, sorghum, flax (linseed) or sugar beet. Furthermore, the oilseed plant may be other Brassicas, cotton, peanut, poppy, mustard, castor bean, sesame, safflower, or nut producing plants. The plant may produce high levels of oil in its fruit, such as olive, oil palm or coconut. Horticultural plants to which the present invention may be applied are lettuce, endive, or vegetable brassicas including cabbage, broccoli, or cauliflower. The present invention may be applied in tobacco, cucurbits, carrot, strawberry, tomato, or pepper.
In a further preferred embodiment, the non-transgenic plant used to produce a transgenic plant of the invention produces oil, especially in the seed, which has less than 20%, less than 10% or less than 5% 18:2 fatty acids and/or which has less than 10% or less than 5% 18:3 fatty acids.
Transgenic plants, as defined in the context of the present invention include plants (as well as parts and cells of said plants) and their progeny which have been genetically modified using recombinant techniques to cause production of at least one polypeptide of the present invention in the desired plant or plant organ. Transgenic plants can be produced using techniques known in the art, such as those generally described in A. Slater et al., Plant Biotechnology - The Genetic Manipulation of Plants, Oxford University Press (2003), and P. Christou and H. Klee, Handbook of Plant Biotechnology, John Wiley and Sons (2004).
In a preferred embodiment, the transgenic plants are homozygous for each and every gene that has been introduced (transgene) so that their progeny do not segregate for the desired phenotype. The transgenic plants may also be heterozygous for the introduced transgene(s), such as, for example, in Fl progeny which have been grown from hybrid seed. Such plants may provide advantages such as hybrid vigour, well known in the art.
The transgenic plants may also comprise additional transgenes involved in the production of LC-PUFAs such as, but not limited to, a Δ6 desaturase, a Δ9 elongase, a Δ8 desaturase, a Δ6 elongase, a Δ5 desaturase with activity on a 20:3 substrate, an omega-desaturase, a Δ9 elongase, a Δ4 desaturase, a Δ7 elongase and/or members of the polyketide synthase pathway. Examples of such enzymes are known in the art and include those described in WO 05/103253 (see, for example, Table 1 of WO 05/103253).
A polynucleotide of the present invention may be expressed constitutively in the transgenic plants during all stages of development. Depending on the use of the plant or plant organs, the polypeptides may be expressed in a stage-specific manner. Furthermore, the polynucleotides may be expressed tissue-specifically.
Regulatory sequences which are known or are found to cause expression of a gene encoding a polypeptide of interest in plants may be used in the present invention. The choice of the regulatory sequences used depends on the target plant and/or target organ of interest. Such regulatory sequences may be obtained from plants or plant viruses, or may be chemically synthesized. Such regulatory sequences are well known to those skilled in the art.
A number of vectors suitable for stable transfection of plant cells or for the establishment of transgenic plants have been described in, e.g., Pouwels et al., Cloning Vectors: A Laboratory Manual, 1985, supp. 1987; Weissbach and Weissbach, Methods for Plant Molecular Biology, Academic Press, 1989; and Gelvin et al., Plant Molecular Biology Manual, Kluwer Academic Publishers, 1990. Typically, plant expression vectors include, for example, one or more cloned plant genes under the transcriptional control of 5' and 3' regulatory sequences and a dominant selectable marker. Such plant expression vectors also can contain a promoter regulatory region (e.g., a regulatory region controlling inducible or constitutive, environmentally- or developmentally-regulated, or cell- or tissue-specific expression), a transcription initiation start site, a ribosome binding site, an RNA processing signal, a transcription termination site, and/or a polyadenylation signal. A number of constitutive promoters that are active in plant cells have been described. Suitable promoters for constitutive expression in plants include, but are not limited to, the cauliflower mosaic virus (CaMV) 35S promoter, the Figwort mosaic virus (FMV) 35 S, the sugarcane bacilliform virus promoter, the commelina yellow mottle virus promoter, the light-inducible promoter from the small subunit of the ribulose-l,5-bis-phosphate carboxylase, the rice cytosolic triosephosphate isomerase promoter, the adenine phosphoribosyltransferase promoter of Arabidopsis, the rice actin 1 gene promoter, the mannopine synthase and octopine synthase promoters, the Adh promoter, the sucrose synthase promoter, the R gene complex promoter, and the chlorophyll α/β binding protein gene promoter. These promoters have been used to create DNA vectors that have been expressed in plants; see, e.g., PCT publication WO 8402913. All of these promoters have been used to create various types of plant- expressible recombinant DNA vectors.
For the purpose of expression in source tissues of the plant, such as the leaf, seed, root or stem, it is preferred that the promoters utilized in the present invention have relatively high expression in these specific tissues. For this purpose, one may choose from a number of promoters for genes with tissue- or cell-specific or - enhanced expression. Examples of such promoters reported in the literature include the chloroplast glutamine synthetase GS2 promoter from pea, the chloroplast fructose- 1,6-biphosphatase promoter from wheat, the nuclear photosynthetic ST-LSl promoter from potato, the serine/threonine kinase promoter and the glucoamylase (CHS) promoter from Arabidopsis thaliana. Also reported to be active in photosynthetically active tissues are the ribulose-l,5-bisphosphate carboxylase promoter from eastern larch (Larix laricind), the promoter for the Cab gene, Cabό, from pine, the promoter for the Cab-1 gene from wheat, the promoter for the Cab-1 gene from spinach, the promoter for the Cab IR gene from rice, the pyruvate, orthophosphate dikinase (PPDK) promoter from Zea mays, the promoter for the tobacco Lhcbl*2 gene, the Arabidopsis thaliana Suc2 sucrose-H30 symporter promoter, and the promoter for the thylakoid membrane protein genes from spinach (PsaD, PsaF, PsaE, PC, FNR, AtpC, AtpD, Cab, RbcS).
Other promoters for the chlorophyll α/β -binding proteins may also be utilized in the present invention, such as the promoters for LhcB gene and PsbP gene from white mustard (Sinapis alba). A variety of plant gene promoters that are regulated in response to environmental, hormonal, chemical, and/or developmental signals, also can be used for expression of RNA-binding protein genes in plant cells, including promoters regulated by (1) heat, (2) light (e.g., pea RbcS-3A promoter, maize RbcS promoter); (3) hormones, such as abscisic acid, (4) wounding (e.g., Wunl); or (5) chemicals, such as methyl jasminate, salicylic acid, steroid hormones, alcohol, Safeners (WO 9706269), or it may also be advantageous to employ (6) organ-specific promoters.
For the purpose of expression in sink tissues of the plant, such as the tuber of the potato plant, the fruit of tomato, or the seed of soybean, canola, cotton, Zea mays, wheat, rice, and barley, it is preferred that the promoters utilized in the present invention have relatively high expression in these specific tissues. A number of promoters for genes with tuber-specific or -enhanced expression are known, including the class I patatin promoter, the promoter for the potato tuber ADPGPP genes, both the large and small subunits, the sucrose synthase promoter, the promoter for the major tuber proteins including the 22 kD protein complexes and proteinase inhibitors, the promoter for the granule bound starch synthase gene (GBSS), and other class I and II patatins promoters. Other promoters can also be used to express a protein in specific tissues, such as seeds or fruits. The promoter for β-conglycinin or other seed-specific promoters such as the napin and phaseolin promoters, can be used. A particularly preferred promoter for Zea mays endosperm expression is the promoter for the glutelin gene from rice, more particularly the Osgt-1 promoter. Examples of promoters suitable for expression in wheat include those promoters for the ADPglucose pyrosynthase (ADPGPP) subunits, the granule bound and other starch synthase, the branching and debranching enzymes, the embryogenesis-abundant proteins, the gliadins, and the glutenins. Examples of such promoters in rice include those promoters for the ADPGPP subunits, the granule bound and other starch synthase, the branching enzymes, the debranching enzymes, sucrose synthases, and the glutelins. A particularly preferred promoter is the promoter for rice glutelin, Osgt- 1 gene. Examples of such promoters for barley include those for the ADPGPP subunits, the granule bound and other starch synthase, the branching enzymes, the debranching enzymes, sucrose synthases, the hordeins, the embryo globulins, and the aleurone specific proteins.
Root specific promoters may also be used. An example of such a promoter is the promoter for the acid chitinase gene. Expression in root tissue could also be accomplished by utilizing the root specific subdomains of the CaMV 35 S promoter that have been identified.
In a particularly preferred embodiment, the promoter directs expression in tissues and organs in which fatty acid and oil biosynthesis take place, particularly in seed cells such as endosperm cells and cells of the developing embryo. Promoters which are suitable are the oilseed rape napin gene promoter (US 5,608,152), the Vicia faba USP promoter (Baumlein et al, 1991), the Arabidopsis oleosin promoter (WO 98/45461), the Phaseolus vulgaris phaseolin promoter (US 5,504,200), the Brassica Bce4 promoter (WO 91/13980) or the legumin B4 promoter (Baumlein et al., 1992), and promoters which lead to the seed-specific expression in monocots such as maize, barley, wheat, rye, rice and the like. Notable promoters which are suitable are the barley Ipt2 or lptl gene promoter (WO 95/15389 and WO 95/23230) or the promoters described in WO 99/16890 (promoters from the barley hordein gene, the rice glutelin gene, the rice oryzin gene, the rice prolamin gene, the wheat gliadin gene, the wheat glutelin gene, the maize zein gene, the oat glutelin gene, the sorghum kasirin gene, the rye secalin gene). Other promoters include those described by Broun et al. (1998) and US 20030159173.
The 5' non-translated leader sequence can be derived from the promoter selected to express the heterologous gene sequence of the polynucleotide of the present invention, and can be specifically modified if desired so as to increase translation of mRNA. For a review of optimizing expression of transgenes, see Koziel et al. (1996). The 5' non-translated regions can also be obtained from plant viral RNAs (Tobacco mosaic virus, Tobacco etch virus, Maize dwarf mosaic virus, Alfalfa mosaic virus, among others) from suitable eukaryotic genes, plant genes (wheat and maize chlorophyll a/b binding protein gene leader), or from a synthetic gene sequence. The present invention is not limited to constructs wherein the non- translated region is derived from the 5' non-translated sequence that accompanies the promoter sequence. The leader sequence could also be derived from an unrelated promoter or coding sequence. Leader sequences useful in context of the present invention comprise the maize Hsp70 leader (U.S. 5,362,865 and U.S. 5,859,347), and the TMV omega element.
The termination of transcription is accomplished by a 3' non-translated DNA sequence operably linked in the chimeric vector to the polynucleotide of interest. The 3' non-translated region of a recombinant DNA molecule contains a polyadenylation signal that functions in plants to cause the addition of adenylate nucleotides to the 3' end of the RNA. The 3' non-translated region can be obtained from various genes that are expressed in plant cells. The nopaline synthase 3' untranslated region, the 3' untranslated region from pea small subunit Rubisco gene, the 3' untranslated region from soybean 7S seed storage protein gene are commonly used in this capacity. The 3' transcribed, non-translated regions containing the polyadenylate signal of Agrobacterium tumor-inducing (Ti) plasmid genes are also suitable.
Four general methods for direct delivery of a gene into cells have been described: (1) chemical methods (Graham et al., 1973); (2) physical methods such as microinjection (Capecchi, 1980); electroporation (see, for example, WO 87/06614, US 5,472,869, 5,384,253, WO 92/09696 and WO 93/21335); and the gene gun (see, for example, US 4,945,050 and US 5,141,131); (3) viral vectors (Clapp, 1993; Lu et al., 1993; Eglitis et al., 1988); and (4) receptor-mediated mechanisms (Curiel et al., 1992; Wagner et al, 1992).
Acceleration methods that may be used include, for example, microprojectile bombardment and the like. One example of a method for delivering transforming nucleic acid molecules to plant cells is microprojectile bombardment. This method has been reviewed by Yang et al., Particle Bombardment Technology for Gene Transfer, Oxford Press, Oxford, England (1994). Non-biological particles (microprojectiles) that may be coated with nucleic acids and delivered into cells by a propelling force. Exemplary particles include those comprised of tungsten, gold, platinum, and the like. A particular advantage of microprojectile bombardment, in addition to it being an effective means of reproducibly transforming monocots, is that neither the isolation of protoplasts, nor the susceptibility of Agrobacterium infection are required. An illustrative embodiment of a method for delivering DNA into Zea mays cells by acceleration is a biolistics α-particle delivery system, that can be used to propel particles coated with DNA through a screen, such as a stainless steel or Nytex screen, onto a filter surface covered with corn cells cultured in suspension. A particle delivery system suitable for use with the present invention is the helium acceleration PDS- 1000/He gun available from Bio-Rad Laboratories.
For the bombardment, cells in suspension may be concentrated on filters. Filters containing the cells to be bombarded are positioned at an appropriate distance below the microprojectile stopping plate. If desired, one or more screens are also positioned between the gun and the cells to be bombarded.
Alternatively, immature embryos or other target cells may be arranged on solid culture medium. The cells to be bombarded are positioned at an appropriate distance below the microprojectile stopping plate. If desired, one or more screens are also positioned between the acceleration device and the cells to be bombarded. Through the use of techniques set forth herein one may obtain up to 1000 or more foci of cells transiently expressing a marker gene. The number of cells in a focus that express the exogenous gene product 48 hours post-bombardment often range from one to ten and average one to three.
In bombardment transformation, one may optimize the pre-bombardment culturing conditions and the bombardment parameters to yield the maximum numbers of stable transformants. Both the physical and biological parameters for bombardment are important in this technology. Physical factors are those that involve manipulating the DNA/microprojectile precipitate or those that affect the flight and velocity of either the macro- or microprojectiles. Biological factors include all steps involved in manipulation of cells before and immediately after bombardment, the osmotic adjustment of target cells to help alleviate the trauma associated with bombardment, and also the nature of the transforming DNA5 such as linearized DNA or intact supercoiled plasmids. It is believed that pre-bombardment manipulations are especially important for successful transformation of immature embryos.
In another alternative embodiment, plastids can be stably transformed. Methods disclosed for plastid transformation in higher plants include particle gun delivery of DNA containing a selectable marker and targeting of the DNA to the plastid genome through homologous recombination (U.S. 5, 451,513, U.S. 5,545,818, U.S. 5,877,402, U.S. 5,932479, and WO 99/05265).
Accordingly, it is contemplated that one may wish to adjust various aspects of the bombardment parameters in small scale studies to fully optimize the conditions. One may particularly wish to adjust physical parameters such as gap distance, flight distance, tissue distance, and helium pressure. One may also minimize the trauma reduction factors by modifying conditions that influence the physiological state of the recipient cells and that may therefore influence transformation and integration efficiencies. For example, the osmotic state, tissue hydration and the subculture stage or cell cycle of the recipient cells may be adjusted for optimum transformation. The execution of other routine adjustments will be known to those of skill in the art in light of the present disclosure.
Agrobacterium-medi&ted transfer is a widely applicable system for introducing genes into plant cells because the DNA can be introduced into whole plant tissues, thereby bypassing the need for regeneration of an intact plant from a protoplast. The use of Agrobacterium-mediatQd plant integrating vectors to introduce DNA into plant cells is well known in the art (see, for example, US 5,177,010, US 5,104,310, US 5,004,863, US 5,159,135). Further, the integration of the T-DNA is a relatively precise process resulting in few rearrangements. The region of DNA to be transferred is defined by the border sequences, and intervening DNA is usually inserted into the plant genome.
Modern Agrobacterium transformation vectors are capable of replication in E. coli as well as Agrobacterium, allowing for convenient manipulations as described (Klee et al., In: Plant DNA Infectious Agents, Hohn and Schell, eds., Springer-Verlag, New York, pp. 179-203 (1985). Moreover, technological advances in vectors for Agrobacterium-mediaXed gene transfer have improved the arrangement of genes and restriction sites in the vectors to facilitate construction of vectors capable of expressing various polypeptide coding genes. The vectors described have convenient multi-linker regions flanked by a promoter and a polyadenylation site for direct expression of inserted polypeptide coding genes and are suitable for present purposes. In addition, Agrobacterium containing both armed and disarmed Ti genes can be used for the transformations. In those plant varieties where Agrobacterium-medistQd transformation is efficient, it is the method of choice because of the facile and defined nature of the gene transfer.
A transgenic plant formed using Agrohacterium transformation methods typically contains a single genetic locus on one chromosome. Such transgenic plants can be referred to as being hemizygous for the added gene. More preferred is a transgenic plant that is homozygous for the added gene; i.e., a transgenic plant that contains two added genes, one gene at the same locus on each chromosome of a chromosome pair. A homozygous transgenic plant can be obtained by sexually mating (selfing) an independent segregant transgenic plant that contains a single added gene, germinating some of the seed produced and analyzing the resulting plants for the gene of interest. I
It is also to be understood that two different transgenic plants can also be mated to produce offspring that contain two independently segregating exogenous genes. Selfing of appropriate progeny can produce plants that are homozygous for both exogenous genes. Back-crossing to a parental plant and out-crossing with a non- transgenic plant are also contemplated, as is vegetative propagation. Descriptions of other breeding methods that are commonly used for different traits and crops can be found in Fehr, In: Breeding Methods for Cultivar Development, Wilcox J. ed., American Society of Agronomy, Madison Wis. (1987).
Transformation of plant protoplasts can be achieved using methods based on calcium phosphate precipitation, polyethylene glycol treatment, electroporation, and combinations of these treatments. Application of these systems to different plant varieties depends upon the ability to regenerate that particular plant strain from protoplasts. Illustrative methods for the regeneration of cereals from protoplasts are described (Fujimura et al, 1985; Toriyama et al, 1986; Abdullah et al., 1986).
Other methods of cell transformation can also be used and include but are not limited to introduction of DNA into plants by direct DNA transfer into pollen, by direct injection of DNA into reproductive organs of a plant, or by direct injection of DNA into the cells of immature embryos followed by the rehydration of desiccated embryos.
The regeneration, development, and cultivation of plants from single plant protoplast transformants or from various transformed explants is well known in the art (Weissbach et al., In: Methods for Plant Molecular Biology, Academic Press, San Diego, Calif., (1988). This regeneration and growth process typically includes the steps of selection of transformed cells, culturing those individualized cells through the usual stages of embryonic development through the rooted plantlet stage. Transgenic embryos and seeds are similarly regenerated. The resulting transgenic rooted shoots are thereafter planted in an appropriate plant growth medium such as soil. The development or regeneration of plants containing the foreign, exogenous gene is well known in the art. Preferably, the regenerated plants are self-pollinated to provide homozygous transgenic plants. Otherwise, pollen obtained from the regenerated plants is crossed to seed-grown plants of agronomically important lines. Conversely, pollen from plants of these important lines is used to pollinate regenerated plants. A transgenic plant of the present invention containing a desired exogenous nucleic acid is cultivated using methods well known to one skilled in the art.
Methods for transforming dicots, primarily by use of Agrobacterium tumefaciens, and obtaining transgenic plants have been published for cotton (U.S. 5,004,863, U.S. 5,159,135, U.S. 5,518,908); soybean (U.S. 5,569,834, U.S. 5,416,011); Brassica (U.S. 5,463,174); peanut (Cheng et al., 1996); and pea (Grant et al., 1995).
Methods for transformation of cereal plants such as wheat and barley for introducing genetic variation into the plant by introduction of an exogenous nucleic acid and for regeneration of plants from protoplasts or immature plant embryos are well known in the art, see for example, Canadian Patent Application No. 2,092,588, Australian Patent Application No 61781/94, Australian Patent No 667939, US Patent No. 6,100,447, International Patent Application PCT/US97/10621, U.S. Patent No. 5,589,617, U.S. Patent No. 6,541,257, and other methods are set out in Patent specification WO99/14314. Preferably, transgenic wheat or barley plants are produced by Agrobacterium tumefaciens mediated transformation procedures. Vectors carrying the desired nucleic acid construct may be introduced into regenerable wheat cells of tissue cultured plants or explants, or suitable plant systems such as protoplasts.
The regenerable wheat cells are preferably from the scutellum of immature embryos, mature embryos, callus derived from these, or the meristematic tissue.
To confirm the presence of the transgenes in transgenic cells and plants, a polymerase chain reaction (PCR) amplification or Southern blot analysis can be performed using methods known to those skilled in the art. Expression products of the transgenes can be detected in any of a variety of ways, depending upon the nature of the product, and include Western blot and enzyme assay. One particularly useful way to quantitate protein expression and to detect replication in different plant tissues is to use a reporter gene, such as GUS. Once transgenic plants have been obtained, they may be grown to produce plant tissues or parts having the desired phenotype. The plant tissue or plant parts, may be harvested, and/or the seed collected. The seed may serve as a source for growing additional plants with tissues or parts having the desired characteristics. Transgenic Hon-Human Animals
A "transgenic non-human animal" refers to an animal, other than a human, that contains a gene construct ("transgene") not found in a wild-type animal of the same species or breed. A "transgene" as referred to herein has the normal meaning in the art of biotechnology and includes a genetic sequence which has been produced or altered by recombinant DNA or RNA technology and which has been introduced into an animal cell. The transgene may include genetic sequences derived from an animal cell. Typically, the transgene has been introduced into the animal by human manipulation such as, for example, by transformation but any method can be used as one of skill in the art recognizes.
Techniques for producing transgenic animals are well known in the art. A useful general textbook on this subject is Houdebine, Transgenic animals - Generation and Use (Harwood Academic, 1997).
Heterologous DNA can be introduced, for example, into fertilized mammalian ova. For instance, totipotent or pluripotent stem cells can be transformed by microinjection, calcium phosphate mediated precipitation, liposome fusion, retroviral infection or other means, the transformed cells are then introduced into the embryo, and the embryo then develops into a transgenic animal. In a highly preferred method, developing embryos are infected with a retrovirus containing the desired DNA, and transgenic animals produced from the infected embryo. In a most preferred method, however, the appropriate DNAs are coinjected into the pronucleus or cytoplasm of embryos, preferably at the single cell stage, and the embryos allowed to develop into mature transgenic animals.
Another method used to produce a transgenic animal involves microinjecting a nucleic acid into pro-nuclear stage eggs by standard methods. Injected eggs are then cultured before transfer into the oviducts of pseudopregnant recipients.
Transgenic animals may also be produced by nuclear transfer technology. Using this method, fibroblasts from donor animals are stably transfected with a plasmid incorporating the coding sequences for a binding domain or binding partner of interest under the control of regulatory sequences. Stable transfectants are then fused to enucleated oocytes, cultured and transferred into female recipients.
Feedstuffs
The present invention includes compositions which can be used as feedstuffs. For purposes of the present invention, "feedstuffs" include any food or preparation for human or animal consumption (including for enteral and/or parenteral consumption) which when taken into the body (a) serve to nourish or build up tissues or supply energy; and/or (b) maintain, restore or support adequate nutritional status or metabolic function. Feedstuffs of the invention include nutritional compositions for babies and/or young children.
Feedstuffs of the invention comprise, for example, a cell of the invention, a plant of the invention, the plant part of the invention, the seed of the invention, an extract of the invention, the product of the method of the invention, the product of the fermentation process of the invention, or a composition along with a suitable carrier(s). The term "carrier" is used in its broadest sense to encompass any component which may or may not have nutritional value. As the skilled addressee will appreciate, the carrier must be suitable for use (or used in a sufficiently low concentration) in a feedstuff such that it does not have deleterious effect on an organism which consumes the feedstuff.
The feedstuff of the present invention comprises an oil, fatty acid ester, or fatty acid produced directly or indirectly by use of the methods, cells or plants disclosed herein. The composition may either be in a solid or liquid form. Additionally, the composition may include edible macronutrients, vitamins, and/or minerals in amounts desired for a particular use. The amounts of these ingredients will vary depending on whether the composition is intended for use with normal individuals or for use with individuals having specialized needs, such as individuals suffering from metabolic disorders and the like.
Examples of suitable carriers with nutritional value include, but are not limited to, macronutrients such as edible fats, carbohydrates and proteins. Examples of such edible fats include, but are not limited to, coconut oil, borage oil, fungal oil, black current oil, soy oil, and mono- and diglycerides. Examples of such carbohydrates include (but are not limited to): glucose, edible lactose, and hydrolyzed search. Additionally, examples of proteins which may be utilized in the nutritional composition of the invention include (but are not limited to) soy proteins, electrodialysed whey, electrodialysed skim milk, milk whey, or the hydrolysates of these proteins.
With respect to vitamins and minerals, the following may be added to the feedstuff compositions of the present invention: calcium, phosphorus, potassium, sodium, chloride, magnesium, manganese, iron, copper, zinc, selenium, iodine, and Vitamins A, E, D, C, and the B complex. Other such vitamins and minerals may also be added.
The components utilized in the feedstuff compositions of the present invention can be of semi-purified or purified origin. By semi-purified or purified is meant a material which has been prepared by purification of a natural material or by de novo synthesis. A feedstuff composition of the present invention may also be added to food even when supplementation of the diet is not required. For example, the composition may be added to food of any type, including (but not limited to): margarine, modified butter, cheeses, milk, yogurt, chocolate, candy, snacks, salad oils, cooking oils, cooking fats, meats, fish and beverages.
The genus Saccharomyces spp is used in both brewing of beer and wine making and also as an agent in baking, particularly bread. Yeast is a major constituent of vegetable extracts. Yeast is also used as an additive in animal feed. It will be apparent that genetically engineered yeast strains can be provided which are adapted to synthesise LC-PUFA as described herein. These yeast strains can then be used in food stuffs and in wine and beer making to provide products which have enhanced fatty acid content.
Additionally, fatty acids produced in accordance with the present invention or host cells transformed to contain and express the subject genes may also be used as animal food supplements to alter an animal's tissue or milk fatty acid composition to one more desirable for human or animal consumption. Examples of such animals include sheep, cattle, horses and the like.
Furthermore, feedstuffs of the invention can be used in aquaculture to increase the levels of fatty acids in fish for human or animal consumption.
Compositions
The present invention also encompasses compositions, particularly pharmaceutical compositions, comprising one or more of the fatty acids and/or resulting oils produced using the methods of the invention.
A pharmaceutical composition may comprise one or more of the fatty acids and/or oils, in combination with a standard, well-known, non-toxic pharmaceutically- acceptable carrier, adjuvant or vehicle such as phosphate-buffered saline, water, ethanol, polyols, vegetable oils, a wetting agent or an emulsion such as a water/oil emulsion. The composition may be in either a liquid or solid form. For example, the composition may be in the form of a tablet, capsule, ingestible liquid or powder, injectible, or topical ointment or cream. Proper fluidity can be maintained, for example, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. Besides such inert diluents, the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfuming agents.
Suspensions, in addition to the active compounds, may comprise suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth or mixtures of these substances.
Solid dosage forms such as tablets and capsules can be prepared using techniques well known in the art. For example, fatty acids produced in accordance with the present invention can be tableted with conventional tablet bases such as lactose, sucrose, and cornstarch in combination with binders such as acacia, cornstarch or gelatin, disintegrating agents such as potato starch or alginic acid, and a lubricant such as stearic acid or magnesium stearate. Capsules can be prepared by incorporating these excipients into a gelatin capsule along with antioxidants and the relevant fatty acid(s).
For intravenous administration, the fatty acids produced in accordance with the present invention or derivatives thereof may be incorporated into commercial formulations.
A typical dosage of a particular fatty acid is from 0.1 mg to 20 g, taken from one to five times per day (up to 100 g daily) and is preferably in the range of from about 10 mg to about 1, 2, 5, or 10 g daily (taken in one or multiple doses). As known in the art, a minimum of about 300 mg/day of fatty acid, especially LC-PUFA, is desirable. However, it will be appreciated that any amount of fatty acid will be beneficial to the subject.
Possible routes of administration of the pharmaceutical compositions of the present invention include, for example, enteral (e.g., oral and rectal) and parenteral. For example, a liquid preparation may be administered orally or rectally. Additionally, a homogenous mixture can be completely dispersed in water, admixed under sterile conditions with physiologically acceptable diluents, preservatives, buffers or propellants to form a spray or inhalant.
The dosage of the composition to be administered to the patient may be determined by one of ordinary skill in the art and depends upon various factors such as weight of the patient, age of the patient, overall health of the patient, past history of the patient, immune status of the patient, etc.
Additionally, the compositions of the present invention may be utilized for cosmetic purposes. It may be added to pre-existing cosmetic compositions such that a mixture is formed or a fatty acid produced according to the subject invention may be used as the sole "active" ingredient in a cosmetic composition.
Production of Oils
Techniques that are routinely practiced in the art can be used to extract, process, and analyze the oils produced by cells, plants, seeds, etc of the instant invention. Typically, plant seeds are cooked, pressed, and extracted to produce crude oil, which is then degummed, refined, bleached, and deodorized. Generally, techniques for crushing seed are known in the art. For example, oilseeds can be tempered by spraying them with water to raise the moisture content to, e.g., 8.5%, and flaked using a smooth roller with a gap setting of 0.23 to 0.27 mm. Depending on the type of seed, water may not be added prior to crushing. Application of heat deactivates enzymes, facilitates further cell rupturing, coalesces the oil droplets, and agglomerates protein particles, all of which facilitate the extraction process.
The majority of the seed oil is released by passage through a screw press. Cakes expelled from the screw press are then solvent extracted, e.g., with hexane, using a heat traced column. Alternatively, crude oil produced by the pressing operation can be passed through a settling tank with a slotted wire drainage top to remove the solids that are expressed with the oil during the pressing operation. The clarified oil can be passed through a plate and frame filter to remove any remaining fine solid particles. If desired, the oil recovered from the extraction process can be combined with the clarified oil to produce a blended crude oil.
Once the solvent is stripped from the crude oil, the pressed and extracted portions are combined and subjected to normal oil processing procedures (i.e., degumming, caustic refining, bleaching, and deodorization). Degumming can be performed by addition of concentrated phosphoric acid to the crude oil to convert non- hydratable phosphatides to a hydratable form, and to chelate minor metals that are present. Gum is separated from the oil by centrifugation. The oil can be refined by addition of a sufficient amount of a sodium hydroxide solution to titrate all of the fatty acids and removing the soaps thus formed.
Deodorization can be performed by heating the oil to 26O0C under vacuum, and slowly introducing steam into the oil at a rate of about 0.1 ml/minute/100 ml of oil. After about 30 minutes of sparging, the oil is allowed to cool under vacuum. The oil is typically transferred to a glass container and flushed with argon before being stored under refrigeration. If the amount of oil is limited, the oil can be placed under vacuum, e.g., in a Parr reactor and heated to 26O0C for the same length of time that it would have been deodorized. This treatment improves the color of the oil and removes a majority of the volatile substances.
Antibodies
The invention also provides monoclonal or polyclonal antibodies to polypeptides of the invention or fragments thereof. Thus, the present invention further provides a process for the production of monoclonal or polyclonal antibodies to polypeptides of the invention. The term "binds specifically" refers to the ability of the antibody to bind to proteins of the present invention but not other known desaturase, conjugase, epoxidase and/or hydroxylase-like polypeptides.
As used herein, the term "epitope" refers to a region of a polypeptide of the invention which is bound by the antibody. An epitope can be administered to an animal to generate antibodies against the epitope, however, antibodies of the present invention preferably specifically bind the epitope region in the context of the entire polypeptide.
If polyclonal antibodies are desired, a selected mammal (e.g., mouse, rabbit, goat, horse, etc.) is immunised with an immunogenic polypeptide such as those provided as SEQ ID NOs: 16 to 30, 73 to 78, 80 and 134. Serum from the immunised animal is collected and treated according to known procedures. If serum containing polyclonal antibodies contains antibodies to other antigens, the polyclonal antibodies can be purified by immunoaffinity chromatography. Techniques for producing and processing polyclonal antisera are known in the art. In order that such antibodies may be made, the invention also provides peptides of the invention or fragments thereof haptenised to another peptide for use as immunogens in animals.
Monoclonal antibodies directed against polypeptides of the invention can also be readily produced by one skilled in the art. The general methodology for making monoclonal antibodies by hybridomas is well known. Immortal antibody-producing cell lines can be created by cell fusion, and also by other techniques such as direct transformation of B lymphocytes with oncogenic DNA, or transfection with Epstein- Barr virus. Panels of monoclonal antibodies produced can be screened for various properties; i.e., for isotype and epitope affinity.
An alternative technique involves screening phage display libraries where, for example the phage express scFv fragments on the surface of their coat with a large variety of complementarity determining regions (CDRs). This technique is well known in the art.
For the purposes of this invention, the term "antibody", unless specified to the contrary, includes fragments of whole antibodies which retain their binding activity for a target antigen. Such fragments include Fv, F(ab') and F(ab')2 fragments, as well as single chain antibodies (scFv). Furthermore, the antibodies and fragments thereof may be humanised antibodies, for example as described in EP-A-239400.
Antibodies of the invention may be bound to a solid support and/or packaged into kits in a suitable container along with suitable reagents, controls, instructions and the like.
In an embodiment, antibodies of the present invention are detectably labeled. Exemplary detectable labels that allow for direct measurement of antibody binding include radiolabels, fluorophores, dyes, magnetic beads, chemiluminescers, colloidal particles, and the like. Examples of labels which permit indirect measurement of binding include enzymes where the substrate may provide for a coloured or fluorescent product. Additional exemplary detectable labels include covalently bound enzymes capable of providing a detectable product signal after addition of suitable substrate. Examples of suitable enzymes for use in conjugates include horseradish peroxidase, alkaline phosphatase, malate dehydrogenase and the like. Where not commercially available, such antibody-enzyme conjugates are readily produced by techniques known to those skilled in the art. Further exemplary detectable labels include biotin, which binds with high affinity to avidin or streptavidin; fluorochromes (e.g., phycobiliproteins, phycoerythrin and allophycocyanins; fluorescein and Texas red), which can be used with a fluorescence activated cell sorter; haptens; and the like. Preferably, the detectable label allows for direct measurement in a plate luminometer, e.g., biotin. Such labeled antibodies can be used in techniques known in the art to detect polypeptides of the invention.
EXAMPLES
Example 1 - Identification of desaturases in the Tribolium castaneum genome
Sixty-four published full length insect desaturase protein sequences available at the end of 2005 were aligned using the Clustal X program (Thompson et al., 1997,). From this sequence alignment, a highly conserved peptide sequence was selected (HNYHHAYPWDYKAAEIGMPLNSTAS LIRLCASLGWAYDLKSV (SEQ ID NO: 31)) and used to search the Tribolium castaneum genome using TBLASTN program (Altschul et al., 1997). From this analysis, 15 desaturase-like sequences were identified which have been termed, Tribdesatl (coding sequence provided as SEQ ID NO:1, amino acid sequence provided as SEQ ID NO:16), Tribdesat2a (coding sequence provided as SEQ ID NO:2, amino acid sequence provided as SEQ ID NO: 17), Tribdesat2b (coding sequence provided as SEQ ID NO:3, amino acid sequence provided as SEQ ID NO: 18), Tribdesat2c (coding sequence provided as SEQ ID NO:4, amino acid sequence provided as SEQ ID NO: 19), Tribdesat3 (coding sequence provided as SEQ ID NO:5, amino acid sequence provided as SEQ ID NO:20), Tribdesat4 (coding sequence provided as SEQ ID NO: 6, amino acid sequence provided as SEQ ID NO:21), Tribdesat5 (coding sequence provided as SEQ ID NO: 7, amino acid sequence provided as SEQ ID NO:22), Tribdesatβa (coding sequence provided as SEQ ID NO:8, amino acid sequence provided as SEQ ID NO:23), Tribdesatόb (coding sequence provided as SEQ ID NO:9, amino acid sequence provided as SEQ ID NO:24), Tribdesat7a (coding sequence provided as SEQ ID NO: 10, amino acid sequence provided as SEQ ID NO:25), Tribdesat7b (coding sequence provided as SEQ ID NO: 11, amino acid sequence provided as SEQ ID NO:26), Tribdesat8 (coding sequence provided as SEQ ID NO: 12, amino acid sequence provided as SEQ ID NO:27), TribdesatlO (coding sequence provided as SEQ ID NO: 13, amino acid sequence provided as SEQ ID NO:28), Tribdesatl l (coding sequence provided as SEQ ID NO: 14, amino acid sequence provided as SEQ ID NO:29) and Tribdesatl2 (coding sequence provided as SEQ ID NO: 15, amino acid sequence provided as SEQ ID NO:30). The sequences listed above as Tribdesat2a, 2b, 2c, 3, 4, 5, 6a, 8, 10, 11, 12 were corrected on the basis of cloning and sequencing of the corresponding cDNAs (see Examples 2-5).
Each of the contigs was then subjected to a gene prediction program in Softberry (http ://www. softberry. com/cgi-bin/programs/gfin/f genesh) . Parameters for Drosophila, Anopheles, C. elegans and Brugia malaya were tested for their ability to predict desaturase genes. For each of the gene predictions, the proteins were subjected to a BLASTP analysis against all proteins in the NCBI database to determine if they resembled desaturases. Neither Anopheles nor Drosophila parameters gave the correct prediction for desaturases except for TribdesatlO which used Anopheles parameters. For some of the desaturases, parameters for predicting genes in C. elegans or B. malaya predicted genes similar to desaturases. For three of the desaturases, none of the predictions using any of the default parameters predicted desaturase genes (Tribdesat4, Tribdesat7a and Tribdesat7b).
For three of the desaturase genes (2 contigs) that could not be predicted using the gene prediction programs, the genomic regions were subjected to a BLASTX comparison with all proteins in the NCBI database. The most 5' region of the genomic fragment resembling desaturases was noted and DNA sequences upstream were translated in silico and visually examined for a methionine residue. This was taken to be the start methionine of the protein sequence. The 3' region of the genomic fragment that resembled desaturases were examined in a similar manner for a stop codon. Each of these protein sequences were visually inspected for all the conserved motifs of desaturases (Table 2). For Tribdesat4, no reasonable prediction of the N- terminus or C-terminus of the protein could be made due to incomplete sequence of the genome in these regions.
Each protein sequence was also examined for the presence of three "histidine boxes" (His boxes) which are motifs that are strongly conserved in all desaturases. The histidine residues in these boxes are thought to be involved in binding iron atoms required for desaturase activity. The amino acid positions and sequences of the His boxes in the proteins and two others from Chauliognathus lugubris (see Example 5) are listed in Table 3, after correction of the predicted protein sequences from analysis of the cDNA clones (Examples 2-5). Table 2 - Presence (V) or absence (X) of conserved desaturase motifs in identified Tribolium amino acid sequences. (Note: Motifs in the sequences may not be identical to the corresponding conserved sequence motif).
Figure imgf000048_0001
Concensus amino acid sequences of motifs: 1, AGAPIRLW (SEQ ID NO: 104); 2, SETDAD (SEQ ID NO: 105); 3, FFFSHVG (SEQ ID NO: 106); 4, QKKY (SEQ ID NO: 107); 5, NSAAH (SEQ ID NO: 108); 6, GEGWHNYHH (SEQ ID NO: 109); 7, PWDY (SEQ ID NO: 110); 8, GWAY (SEQ ID NO: 111). Table 3. Amino acid location and sequences of histidine boxes in Triboliwn and soldier beetle desaturase sequences. The number for each box corresponds to the location in the protein of the first amino acid residue of the motif.
Figure imgf000049_0001
Example 2 - Cloning of Tribdesat4
The inventors attempted to obtain Tribdesat4 using inverse PCR on cDNA as we could not predict from the genome sequence either the beginning or the end of the gene. First strand synthesis was performed by mixing 3 μl larval RNA (24 μg), 100 pmol of each of the oligonucleotide primers from Clontech, Smart IV Oligo 5'AAGCAGTGGTATCAACGCAGAGTGGCCATTACGGCCGGG-S' (SEQ ID NO: 32); and CDS/3' (5'ATTCTAGAGGCCGAGGCGGCCGACATG-d(T)30VN) (N=A, G, C or T; V=A, G or C (SEQ ID NO: 33)). This was incubated at 72oC for 2 min after which it was immediately cooled on ice. To the contents of the tube was added 2 μl 5X first strand buffer (Powerscript; Clontech), lμl DTT (20 mM), lμl dNTP mix (final concentration of 200μM each dNTP) and 1 μl Powerscript reverse transcriptase (Clontech). First strand cDNA synthesis was allowed to occur at 42oC for 1 hour, after which it was immediately cooled on ice. NaOH (1 μl, 25 mM) was added and incubated at 68oC for 30 min. An aliquot of the first strand synthesis (6 μl) was mixed with 5 μl 1OX Advantage 2 Buffer (Clontech), lμl 5OX dNTP mix, lμl CDS/5 oligo (5 'AAGCAGTGGTATCAACGCAGAGT; Clontech (SEQ ID NO: 34)), lμl CDS/3 oligo, lμl 50X Advantage 2 Polymerase and made to a final volume of 50μl. The mixture then went through a temperature regime as follows: 72°C/10 min; 95°C/1 min; then 3 cycles of 95°C/15 sec, 68°C/8 min.
An aliquot (5 μl) was separated on a 1% agarose gel to ensure that amplification had occurred. The sample was then digested with Sfil for 2 hours at 50oC and the restriction enzyme removed using the QIAgen PCR purification kit according to the manufacturer's instructions. The eluted sample was diluted for ligation to ensure intramolecular ligations rather than intermolecular ligation. Digested DNA (0.5 μg) was diluted in a ligation mixture of 500μl and incubated overnight at 16oC. The ligation was concentrated by glycogen precipitation (150 μg glycogen, 1.4 ml ice-cold 95% ethanol at -80oC for 2 hours) after which DNA was pelleted by centrifugation (12 000g, 20 min). The DNA was resuspended in lOμl sterile water. The inverse PCR reaction was performed on 1 μl of this precipitated DNA and included 5μl 1OX Advantage 2 PCR Buffer (Clontech), 10 pmol Desat4 GSPl (5'ATTATGAGCGGATCGGCTTCCAAGTC (SEQ ID NO: 35), 10 pmol Desat4 GSP2 (5'CGAAACAATTTGGAATTCGTTTTGGG (SEQ ID NO: 36), 200μM each dNTP, lμl 5OX BD Advantage 2 Polymerase and sterile water to a final volume of 50μl. The PCR conditions were as follows: 95oC/l min, then 30 cycles of 95oC/30 sec, 68oC/3 min, then 68oC/3 min.
An aliquot (5μl) of the PCR was separated on a 1% agarose gel. A single band was noted and cloned into pGEM-T-Easy. The insert was sequenced and shown to contain most of the 5' end of Tribdesat4 and the entire 3' end of Tribdesat4. The existing 5' end of Tribdesat4 was then subject to BLASTN analysis against available sequences of ESTs from T. castaneum. One EST was identified from this analysis (Accession No. DT795463) and the rest of the 5' end was identified.
Primers were designed to the 5' and 3' end of the coding region of Tribdesat4 and used in an RT-PCR reaction to obtain full length Tribdesat4. The RT-PCR reaction consisted of 25 μl 2X Reaction mix (Invitrogen), 37 ng RNA, 1 μl Superscript II RT/Platinum Taq DNA polymerase (Invitrogen), 100 pmol of each primer and sterile water to 50 μl. The reaction conditions were as follows: 50oC/30 min, 94oC/2 min, then 35 cycles of 94oC/30 sec, 55oC/l min, 72oC/l min, then 72oC/5 min. An aliquot was run on a 1% agarose gel. A single band (approximately lkb) was obtained and cloned into pGEM T Easy. The sequence of the insert was examined and verified as the full length cDNA. SEQ ID NO: 6 shows the nucleotide sequence of the coding region of the cDNA.
Example 3 - Amplification of desaturase sequences from adult Tribolium RNA
RNA was extracted from T. castaneum (strain TC4) beetles using the Trizol method (Invitrogen). 100 mg of adult beetles were homogenized in 1 ml Trizol reagent and the mixture incubated for 5 minutes at 250C, after which 200 μl chloroform was added to the sample. The mixture was shaken by hand for 15 seconds and then allowed to settle for 3 minutes at 250C. The organic and aqueous layers were separated by centrifugation (12,000g, 15 minutes, 4°C). The upper aqueous layer was transferred to a fresh tube and the RNA precipitated by the addition of 500 μl isopropanol. After 10 minute incubation at 250C, the RNA was pelleted by centrifugation at 12,000g/10 minutes at 4°C. The RNA pellet was washed once with 75% ethanol and then air-dried for 10 minutes. The RNA was then dissolved in 30 μl of RNase-free water. There was a noticeable brown colouration to the RNA which was further purified using the RNeasy mini protocol for RNA cleanup (QIAgen), according to the manufacturer's instructions.
RT-PCR amplification reactions were carried out for 10 desaturase sequences (Tribdesatl, 2a, 2b, 3, 5, 6a, 6b, 8, 10 and 11). To each sample was added 25 μl 2X Reaction mix (Invitrogen), 37 ng RNA, 1 μl Superscript II RT/Platinum Taq DNA polymerase (Invitrogen), 100 pmol of each primer and sterile water to 50 μl. The reaction conditions were as follows: 50°C/30 min, 94°C/2 min, then 35 cycles of 94°C/30 sec, 55°C/1 min, 72°C/1 min, then 72°C/5 min (PCR conditions 55/72).
An aliquot of each sample was run on a 1% agarose gel. Those samples containing a single amplification product of about lkb (Tribdesatl, 3, 5, 6a, 10, 11) were purified using the QIAquick PCR purification kit (QIAgen) and cloned into pGEM T Easy (Promega). Ligation mixtures were transformed into E. coli DHlOB using standard molecular biological techniques (Sambrook et al., 1989). The DNA sequence of clones containing inserts was examined. Comparisons of gene predictions with those of RT-PCR products obtained for Tribedesat2b, 3, 6b, 10 and 11 are shown in Figures 1 to 5. It was apparent that the sequences as predicted from the genome sequence were not the same as the observed cDNA sequences, in some cases this would have been due to incorrect predictions for coding sequences from the genomic sequence. In other cases, differences such as single nucleotide polymorphisms may have been due to different alleles of the genes present in populations of the insects.
RNA was also extracted from T. castaneum larvae using the Trizol method as above. RT-PCR reactions were carried out to amplify 6 desaturase sequences (Tribdesat2a, 2b, 2c, 6b, 8 and 12) by the method see as above. No amplification products were observed after a first round of PCR and so the reactions were subject to a second round of PCR. Amplification products of about 1 kb were observed in the Tribdesat2b, Tribdesat2c, Tribdesatl2 and Tribdesat8 samples. These products were extracted from an agarose gel using the QIAquick gel extraction kit (QIAgen) and cloned into pGEM T Easy (Promega). The DNA sequences of clones containing inserts were examined. Comparisons of gene predictions with those of RT-PCR products obtained for Tribdesat2b and 6b are shown in Figures 1 and 3, respectively.
Example 4. Amplification of desaturase gene fragments from Soldier Beetles (Chauliognathus lugubris)
Amplification of internal desaturase fragments from male Soldier Beetles RNA was extracted from 100 mg of adult male C. lugubris beetles using the Trizol method as described above for Tribolium. The synthesis of cDNA was performed using the Invitrogen Superscript II and using a polyT primer (5'TTTTTTTTTTTTTTTTTT (SEQ ID NO: 81)). The polyT primer (100 pmol), RNA (2 μl) and RNase-free water to a final volume of 15.5 μl were incubated at 7O0C for 10 minutes and then chilled on ice. To this was added 1 OXPCR Buffer (2.5 μl), 25 mM MgCl2 (2.5 μl), 10 mM dNTP mix (1 μl) and 0.1M DTT (2.5 μl). This mixture was heated to 420C for 1 minute after which Superscript II Reverse Transcriptase (1 μl) was added and the mixture left for 50 minutes at 420C. Superscript II Reverse Transcriptase was inactivated at 7O0C for 15 minutes and RNA degraded with lμl RNaseH (2 Units, 30 minutes at 370C).
PCR reactions were setup containing 1 μl dNTPs (200 μM each dNTP), 5 μl 10x ThermoPol Buffer (NEB), 1.5 μl F2 (5^TTCTTCTTCKCNCAYKTHGGNTGG (SEQ ID NO: 82)), 1.5 μl R2 (5TGRTGGTAGTTGTGVHANCCCTC (SEQ ID NO: 83)), 5 μl cDNA, 0.1 U Taq DNA Polymerase (NEB) and sterile water to 50 μl. The PCR conditions were as follows: an initial denaturation of 940C for 3 minutes and then 30 cycles of 94°C/15s, 48°C/30s, 72°C/2 min and then a final extension of 72°C/5 min. An aliquot was separated on a 1.5% agarose gel and a band of approximately 400 bp was visualised. The PCR reaction was purified using the QIAgen QIAquick PCR purification kit according to the manufacturer's instructions and cloned into pGEM T Easy (Promgea). Approximately 30 clones containing inserts were examined by DNA sequence analysis. A total of three unique desaturase sequences were identified corresponding to CL6 (SEQ ID NO: 75), CL7 (SEQ ID NO: 76) and CL8 (SEQ ID NO: 77).
Amplification of internal desaturase fragments from female Soldier Beetles RNA was extracted and cDNA synthesised as described above. Two degenerate PCRs were performed on female adult RNA using the degenerate primers F2/R2 and Clu_f (5OCNCAYMGNYTNTGGGCNCA (SEQ ID NO: 84)) / Clu-r (5^AANRYRTGRTGGTAGTTGIG (SEQ ID NO: 85)) (see Table 4) by the same method as above. Amplification products of approximately 400 bp (F2/R2) and 550 bp (Clu_f/Clu_r) were visualised and cloned into pGEM T Easy. Each insert from 70 colonies from the F2/R2 transformation was amplified by PCR using the T7 (5'TAATACGACTCACTATAGGG (SEQ ID NO: 86)) and SP6 (5^ATTTAGGTGACACTATAG (SEQ ID NO: 87)) primers and analysed. Since approximately 70% of the sequenced clones from male soldier beetle tissue that had sequence similarity with desaturases were CL6, the PCR products were digested with restriction enzyme Rsal to remove CL6 clones. Of the 70 PCR products obtained, only 20 were not digested with Rsal. Resultant desaturase fragments obtained were CL7, CL8 and a new unique desaturase fragment called CL9.
Sixteen further clones were obtained from another degenerate PCR reaction using female adult RNA and primers Clu_f/Clu_r, and their nucleotide sequences obtained and compared to the previous clones. One of these was designated CLl .
Table 4. The tissue source of RNA and degenerate primer pairs used to isolate internal desaturase fragments from C. lugubris.
Figure imgf000053_0001
Figure imgf000054_0001
Amplification of degenerate PCR from defence gland tissue A mixed population of males and females were dissected to obtain their defence gland tissue. Initially, the heads and the elytra were removed. The remaining tissue was divided into two parts - the abdomen sample and the defence gland tissue.
RNA was extracted and cDNA synthesised as described above. Degenerate PCR was performed with three different combinations of primers. These were Clu_f/Clu_r, F2/Clu_r and XRP2b (5TTYTTYTWYKCNCAYATGGGNTGG (SEQ ID NO: 88))/Clu_r. After analysis on a 1.5% agarose gel, a band of the expected size was observed only in the sample with the XRF2b/Clu_r combination. No amplification products were observed with either of the other two pairs even after 60 cycles of PCR. The PCR products obtained were purified using the QIAgen QIAquick PCR purification kit and cloned into pGEM-T-Easy. Eleven clones were examined by sequence analysis. New desaturase gene fragments were obtained and designated CL3 and CL5. The CL5 internal fragment appeared to contain remnants of an intron, as determined by a section that did not show close sequence similarity with desaturases and also affected the reading frame, thereby inserting a stop codon.
5 " and 3 * RACE to obtain full length sequence information Primers for 5'- and 3'-RACE (Table 5) were designed based on sequences internal to the degenerate primer binding sites for each of the clones described above.
Table 5. Primers (5' to 3') used for 5^ and 3' RACE.
Figure imgf000054_0002
The Clontech Creator™ SMART™ System was used to obtain both 5Λ and 3^ RACE products according to the manufacturer's instructions with RNA from either abdomen or defence gland tissue. Oligonucleotide primers Smart IV (5^AAGCAGTGGTATCAACGCAGAGTGGCCATTACGGCCGGG (SEQ ID NO: 97)) and CDS III/3Λ (S'ATTCTAGAGGCCGAGGCGGCCGACATG-dCrboN^N; N=A5G5C or T; N-1=A5G or C (SEQ ID NO: 98)) were used for the reverse transcription-PCR. The 5' RACE reactions used primer CDS V 5"AAGCAGTGGTATCAACGCAGAGT (SEQ ID NO: 99), and CLXfront primer. The 3" RACE reactions used primers CDS III/3" PCR and CLXend.
Clones were screened by colony PCR for insertions. Any clones that gave a band of the expected size (~350 bp) were examined by DNA sequence analysis. The DNA sequence was initially examined for sequence similarity with desaturases. Full length sequences were compiled in silico using sequence information from internal sequence, 5ΕACE and 3'RACE products and examined for an open reading frame beginning with an ATG and ending in a stop codon that when translated possessed all the motifs of desaturases (Table 2).
Example 5. Isolation of full length C. ϊusubris desaturase cDNAs
Full length cDNA clones of CLl and other desaturase-like sequences were obtained using a one-step RT-PCR kit from Invitrogen according to the manufacturer's instructions and PCR conditions 55/72 (above). The primers used were GGTACCATGCCACCCAACGCCCAC (SEQ ID NO: 100) and GAATTCTCACTTTTGTTTGTGAGT (SEQ ID NO: 101). Bands of approximately 1 kb were purified using the QIAquick PCR purification kit from QIAgen according to the manufacturer's instructions and cloned into pGEM-T-Easy. Positive clones were obtained and examined by DNA sequence analysis. After verification of sequence, the pGEM-T-Easy clones containing the desaturase cDNAs were digested with restriction enzymes to excise the inserts, separated on a 1% agarose gel and the ~lkb fragments were excised and purified using the QIAquick gel extraction kit (QIAgen) for cloning into yeast expression vectors such as pYES2 (below).
Example 6. Isolation of desaturase genes from Chauliosnathus nohiliatus
Chaulioenathus nobiliatus (Erichson) is another species of soldier beetle, which resembles C. lugubris but is generally smaller in size and less prevalent in the south-east regions of Australia. These beetle species are not closely related to moths such as Tribolium, indeed they are widely separated within the Insectae.
RNA was extracted from C. nobiliatus adults using the Trizol method as above. cDNA was synthesised using Superscript II kit from Invitrogen. Three degenerate PCRs were performed. The first used the degenerate primers F2/Clu_r, the second used the degenerate primers XRF2b/Clu_r and the third with degenerate primers Clu_f/Clu_r. An aliquot of each reaction was separated on a 1.5% agarose gel and a band was observed for the first two reactions. These were purified using the QIAquick PCR purification kit (QIAgen) and cloned into pGEM-T-Easy. A total of 12 clones from the first PCR and 14 from the second PCR were examined by DNA sequence analysis. Three unique desaturase sequences were obtained and their corresponding orthologues from C. lugubris as well as the signature motif as described by Knipple et al. (2002) are shown in Table 6.
Table 6. The internal desaturase fragments isolated from C. nohϊliatus and their corresponding orthologues in C. lugubris. Also shown is the signature motif and the primers used to isolate these internal fragments.
Figure imgf000056_0001
A second approach used to isolate internal desaturase fragments from C. nobiliatus was to use primers specific to C. lugubris desaturases. The cDNA synthesised as described above was used as a template for PCR with specific primers to CL6, CL7, CL9, and CLl. An aliquot of each reaction was separated on a 1.5% agarose gel and bands were observed with the CL7, CL9 and CLl -specific primers but no products were observed for either the CL6 or the CLIO primers. The CL7, CL9 and CLl products were purified using the QIAgen QIAquick PCR purification kit and cloned into pGEM-T-Easy. Positive clones were obtained and examined by DNA sequence analysis.
Full-length cDNA sequences for the CL3 and CLl orthologues were amplified from C. nobiliatus RNA using the One-step Superscript II Reverse Transcriptase/Platinum Taq DNA Polymerase kit from Invitrogen as described in Example 5. The primers used are shown in Table 7. A clone obtained from each reaction was sequenced to confirm identity of each cDNA. Table 7. Primers used to obtain full length C. nobiliatus orthologues of selected desaturases.
Figure imgf000057_0001
Example 7. Isolation of desaturase genes from cDNA libraries
Construction of a cDNA library from C. lugubris abdomen tissue RNA was extracted from C. lugubris abdomen tissue using the method described above. A cDNA library was constructed using the Invitrogen Cloneminer kit according to the manufacturer's instructions. A fraction of the library was transformed into E. coli DHlOB ElectroMax competent cells (Invitrogen) and transformants selected on LB agar plates containing 25 μg/ml kanamycin. Approximately 90% of the clones contained inserts with an average insert size of 1.5 kb. Desaturase clones were identified and isolated from this library by PCR methods or by hybridisation screening using probes derived from the clones described above.
PCR method for screening plasmid cDNA library
A volume of plasmid cDNA library representing approximately 50 colony forming units was seeded into 96 well plates containing 300μl LB supplemented with 50μg/ml kanamycin and grown overnight at 370C with shaking. lOOμl of each liquid culture from the wells across the plate were pooled into 1.5ml tubes to yield eight "lane pools" and a plasmid extraction was performed using a QIAquick miniprep spin kit (Qiagen) according to manufacturers instructions. lOμl of each "lane pool" elutant was mixed into a 1.5ml tube creating a "plate pool". PCR screening was done on each "plate pool" using gene specific primers and run out on 1% TAE agarose gel to identify positive plate pool/s. Once positive plate/s were identified PCR screening was repeated on each "lane pool" corresponding to a positive "plate pool" and run out on 1% TAE agarose gel to identify positive "lane pools", and in subsequent screening rounds on single wells and individual colonies to isolate positive clones. Plasmids which contained the gene of interest were analysed by restriction enzyme digest and sequence analysis.
Screening using non-radioactive colony hybridisation.
10-fold serial dilutions of the cDNA library were made (1-106) and plated on LB agar supplemented with 50μl/ml kanamycin to determine the dilution which produced approximately 300-500 colonies per 82mm petri dish. Transformed bacteria of a suitable dilution were spread on LB agar plates supplemented with 50μg/ml kanamycin and incubated overnight at 370C. Nylon membranes (Hybond-N+, Amersham Biosciences) were used to obtain colony lifts and treated according to standard methods for colony hybridisation. Biotin labelled DNA probes were prepared using desaturase gene fragments labelled using dNTPs plus Biotin-dATP and DNA polymerase I Klenow fragment according to NEBlot phototope kit (New England Biolabs). Probes were hybridised to the membranes overnight at temperatures of 650C (high stringency) or 550C (moderate stringency). The hybridised biotinylated DNA was detected according to the Phototope-Star detection Kit manual (New England Biolabs) following the modification for colony hybridisations detailed in Appendix C. The membrane was exposed to Hyperfϊlm ECL (Amersham Biosciences) for 1 minute and processed manually according to Amersham Biosciences recommendations. Positively hybridising clones were analysed by restriction enzyme digest and sequence analysis.
Example 8. Yeast expression vector construction and functional analysis of genes
Each of the full-length desaturase genes was expressed in Saccharomyces cerevisiae for functional characterization using the yeast expression vectors pYES2 or pVT100-U (Vernet, T et al. 1987). Each of the genomic regions containing putative desaturase genes was examined for restriction nuclease recognition sites. Restriction enzymes were chosen that did not cut the genomic regions but would facilitate directional cloning into pYES2 or pVT100-U. Primers were designed (Table 8) to the most 5' and 3' regions of the predicted desaturase genes with restriction sites at the 5' end of each of the primers to allow directional cloning.
DNAs of the yeast vectors were prepared and digested with the corresponding restriction enzymes and then dephosphorylated using Calf Intestinal Alkaline Phosphatase (Roche). After dephosphorylation, the digested vectors were purified using the QIAquick PCR purification kit (QIAgen). Gel extracted desaturase- containing fragments were ligated with digested vector and ligations transformed into E. coli DHlOB with transformants selected on LB agar plates with 100 μg/ml ampicillin. Clones containing inserts were confirmed by restriction enzyme analysis.
Table 8 - Primers used for directional cloning into pYES2. Restriction sites are shown in bold face.
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Transformation of expression constructs into Saccharomyces cerevisiae
Two S. cerevisiae host strains were used as recipients for pYES-derived constructs. These were S288C (genotype MATα, SUC2 gal2 mal mel flol flo8-l hapl (Mortimer and Johnston (1986)) and OLEl (his3Δl, leu2Δ0, ura3Δ0, YMR272c::kanMX4) which is mutant for the gene encoding Δ9-desaturase and could be used for complementation analysis. Five S. cerevisiae host strains were used as recipients for ρVT-100 derived constructs. These were S288C, OLEl, and INVSCi (MATa, his3Dl Ieu2 trpl-289 ura3-52), YPH499 (MATa, ura3-52 lys2-801_amber ade2-101_ochre trpl-Δ63 hisl-Δ200 Ieu2-Δl) and ELO-I (MATa his3Δl leu2Δ0 metl5Δ0 ura3Δ0 ΔELO1). These strains were treated the same during transformation except that 17:1 (cis-10-heptadecenoic acid, 1 mM) and tergitol (NP-40; 1%) were added in all media in which S. cerevisiae OLEl was grown and geneticin (200μg/ml) was added to all media in which ELO-I was grown.
For transformation, each S. cerevisiae strain was streaked onto YPD (20g/l peptone, lOg/1 yeast extract, 2% glucose) and grown for several days at 3O0C. Transformations were performed using the Sigma Yeast Transformation kit according to manufacturer's instructions. Transformants were selected on SCMM-U agar plates (6.7 g/1 yeast nitrogen base without amino acids, 1.92 g/1 yeast synthetic drop-out media supplement without uracil, 2% glucose and 2% agar) at 300C for up to 5 days. A number of transformants were selected for each construct and tested for the presence of plasmid DNA.
Confirmation of transformants
To confirm the identity of the expression constructs within transformants, DNA was isolated from a loopful of growth using the Y-DER yeast DNA extraction kit from Pierce (Illinois) according to the manufacturer's instructions. The presence of the plasmid was analysed and confirmed by PCR analysis using a gene specific reverse primer and a T7 primer (vector primer). Positive bands were observed and the corresponding transformants were retained for further analysis. Assessment of OLEl complementation
Fifty microlitres of OLEl transformants grown on SCMM-U containing cis- 10-heptadecenoic acid (1 mM) and 1% tergitol was pelleted by centrifugation (13,000rpm, 3 ses) and resuspended in 50 μl YP (20g/l peptone, lOg/1 yeast extract). The resuspended mixture was diluted 1:10 and 20 μl of each mixture was spotted onto YP containing 2% galactose plates to induce desaturase gene expression and these were incubated for 3-5 days at 30°C. The presence or absence of growth was noted. Control plates containing cis-10-heptadecenoic acid (1 mM) and 1% tergitol were also used to ensure yeast viability. The results for the OLEl complementation tests for Tribolium and Chaulioganthus desaturases were as follows. Positive complementation was observed for Tribdesat2a, Tribdesat5, Tribdesatβa and Tribdesatl l, the last one with a lag phase. Possibly weak complementation was observed for Tribdesat2b and Tribdesatδ. No complementation was observed for Tribdesatl, Tribdesat2c, Tribdesat4, TribdesatlO, Tribdesatl2, CLl and CL3.
Complementation of the yeast OLEl phenotype occurs when a heterologously expressed gene expresses a functional Δ9- or Δl l -desaturase and results in the production of palmitoleic, oleic, or cis-11-octadecanoic fatty acids. Therefore, it was concluded that Tribdesat2a, Tribdesat5, Tribdesatόa and Tribdesatl 1 were desaturases able to desaturate the native lipids of OLEl yeast cells to produce one of the complementing fatty acids and therefore were likely to be Δ9 desaturases active on C16:0 or C18:0, or Δll desaturases active on C18:0, whereas Tribdesatl, Tribdesat2c, Tribdesat4, TribdesatlO, Tribdesatl2, CLl and CL3 were unlikely to encode one of these activities and therefore likely to encode a different desaturase.
Growth of transformed S. cerevisiae for substrate feeding experiments Yeast transformants derived from stains S288C or OLEl were inoculated into 25 ml of synthetic minimal defined medium for yeast without Uracil (SCMM-U medium) containing 2% glucose and additionally 0.5 mM cis-11-heptadecenoic acid for the OLEl strain. This inoculation culture was grown for 24 - 48 hr at 30°C with shaking. The OD600 of the culture was determined. From this, the amount of culture necessary to obtain an OD600 of 0.4 in 50ml of induction medium was calculated. This amount of inoculation culture was removed and the cells pelleted at 1500 x g for 5 minutes at 40C. The cells were then resuspended in 10 ml of induction medium, SCMM-U containing 2% galactose, 1% raffmose and 0.5 mM fatty acid substrate (added in ethanol/20% tergitol) and additionally 0.5 mM cis-11-heptadecenoic acid for OLEl derived transformants. Each culture was grown for 24 - 48 h at 3O0C with shaking. Either 2 ml or the total culture was then used for analysis of fatty acids after harvesting cells by centrifugation, storage at -200C if needed. Extraction of lipids from transformed S. cerevisiae and analysis
The yeast cells from fatty acid feeding experiments were washed in 1% tergitol in water and pelleted at 1500 x g for 5 minutes at 4°C. Cell pellets (200-500mg) were then resuspended in water, transferred to glass test tubes and the cells again pelleted. Water was removed from the cell pellets in a Savant SpeedVac Plus SCIlOA concentrator/dryer. Each pellet was used either used directly or lipids were extracted with solvent as described below.
Lipids were extracted based on a Modified Folch Method (Protocol 7, pp22-24, Lipid Analysis, Hamilton and Hamilton, 1992). Lipids were extracted with 2 ml chloroform/methanol (2:1), and 0.5 ml of a saline solution, 0.9% w/v NaCl in water, was added. This was mixed thoroughly by vortexing. The layers were then allowed to separate; when a large amount of cell debris was present the samples were centrifuged at 2500 x g for 5 minutes. The top aqueous layer was removed and discarded. The bottom solvent layer was removed to a clean glass test tube. This was then dried down under nitrogen at 30°C.
Lipids were then derivatised prior to analysis by two different methylation methods. The first was the basic methylation of incorporated fatty acids, i.e. those found in triacyl glycerides or phospholipids, not free fatty acids, based on the method of Christie (2003). The lipid sample was first dissolved in 0.5 ml hexane. Methyl acetate, 60 μl, was added, followed by 50 μl sodium methoxide at 0.5 M in methanol. The sealed test tube was then mixed thoroughly and placed in a heating block at 50°C for 10 minutes. After being allowed to cool for 5 minutes, a drop of acetic acid was added. The sample was dried in a gentle stream of nitrogen at 30°C, dissolved in 200 μl hexane and transferred to a vial prior to GC analysis.
The second method was an acidic methylation of all free and incorporated fatty acids, based on the method of Lewis et al. (2000). A methanolic solution, 2 ml of methanol/hydrochloric acid/ chloroform (10:1:1), was added to the dried yeast pellet or extracted lipid sample. The sealed test tube was mixed thoroughly and placed in a heating block at 90°C for 60 minutes. After being allowed to cool for 10 minutes, 1 ml of 0.9% saline (NaCl in water) was added. The methylated fatty acids were then extracted with 0.3 ml hexane. The hexane layer was transferred to a vial prior to analysis.
These fatty acid methyl ester (FAME) samples were analysed by GC and GCMS. GC analysis was carried out with a Varian 3800 gas chromatograph fitted with a flame ionisation detector (FID) and a BPX70 capillary column (length 30m, i.d. 0.32mm, film thickness 0.25 μm). Injections were made in the split mode using helium as the carrier gas and an initial column temperature of 100°C. The temperature was raised at 3°C/minute until 150°C, then raised at 5°C/minute until 170°C, held for 5 minutes, then raised at 50°C/minute until 2550C. GC/MS analysis was carried out under similar chromatography conditions but with an initial column temperature of 60°C, raised at 20°C/minute until 170°C, held for 5 minutes, then raised at 50°C/minute until 2550C. Detection was carried out using either a TEC PolarisQ Ion Trap or a Varian 1200 Single Quadrupole mass spectrometer. Mass spectra were acquired under positive electron impact in full scan mode between 50 - 400 aniu at the rate of 2 scans per sec. The mass spectra corresponding to each peal- in the chromatogram was automatically compared with spectra in the computerised NIST library. Test spectra that matched library spectra with a high degree of accuracy and eluted at the same time as an authentic standard or eluted at a plausible retention time, were tentatively identified. Confirmation of the identity of a fatty acid was achieved by the conversion of the fatty acid to its dimethyloxazoline (DMOX) derivative using the method of Yu et al. (1988) and comparison with DMOX mass spectra described in Dobson and Christie (2002) and references within.
Results of functional analysis in yeast.
From the experiments carried out as described above and summarised in Table 10 (below), it was concluded as follows:
Tribdesat2a had Δ9 desaturase activity on saturated fatty acid carbon chain length from C10:0 to C16:0 when these fatty acids were fed to the yeast transformants. Activity was greatest on 14:0, but 15:0 and 16:0 were also efficient substrates. It had no detectable desaturase activity on 18:0, 20:0 or even longer saturated fatty acids in the yeast cells. The protein had a predicted size of 320 amino acids and included the 8 conserved desaturase motifs and three His boxes (Table 3). On the basis of the sequence homology with known acyl-CoA desaturases, this enzyme was presumed to be acting on the acyl-CoA substrate. This enzyme was therefore characterised as a myristoyl-CoA Δ9 desaturase.
Tnbdesatlb and Trϊbdesatlc were closely related proteins, having 64% amino acid sequence identity based on global alignment using BLOSUM62, which catalysed production of 5-hexadecenoic and 5-octadecenoic acids from palmitic acid and stearic acid, respectively. They were therefore characterised as Δ5 desaturases acting on the saturated substrates C16:0 and C18:0. The efficiency of conversion in yeast cells of C18:0 to C18:1A5 was more than twice as efficient as the conversion of C16:0 to C16:1Δ5 (Table 9). The enzymes were able to convert at least 8.0% of the substrate to the product in each case, to at least 40% for Tribdesat2b on C 18:0. The enzymes did not detectably desaturate substrates of C14 or shorter when fed to the yeast cells, including C14:0 or C14:1Δ9, or the substrates of C20 or longer including C20:0 or C20:lΔ13. Furthermore, the enzymes did not desaturate monounsaturated or polyunsaturated fatty acids including C16:1Δ9, C18:1Δ9, C18:2 or C18:3. The proteins included the 8 conserved desaturase motifs and three His boxes (Table 3) and showed sequence homology with known acyl-CoA desaturases. They were therefore characterised as stearoyl-CoA Δ5 desaturases. They also have palmitoyl-CoA Δ5 desaturase activity.
Table 9. Fatty acid composition (% total fatty acids) of yeast cells expressing Tribdesat2b, or empty pYes vector, supplied with 0.5mM 18:0.
Beetle desaturase Fatty add composition (o/o total fatty acids) 12:0 14:0 15:0 16:0 16:1Δ5 17:0 17:1 18:0 18:1Δ5
TD2b + 18:0 2.4 0.7 0.5 24.3 3.2 0.2 62 .4 3 .2 3.0 p Yes empty + 18:0 1.6 0.4 0.4 22.7 O 0.4 65 .7 8 .9 0
Cahoon et al. (2000) isolated a gene from Limnanthes (meadowfoam) which encoded a Δ5 desaturase which was more active on C20:0 than C18:0 or C16:0 and is therefore was not a stearoyl-CoA Δ5 desaturase as defined herein. Sayanova et al. (2007) cloned two genes encoding Δ5 desaturases active on acyl-CoA substrates from Anemone leveillei seeds. The desaturase AL21 was active on both saturated and unsaturated substrates, Cl 6:0, Cl 8:0 and C20:2, while ALlO was active only on C20:2, n-6. However, AL21 was more active on the unsaturated substrate than the saturated substrate so it would not be considered as a stearoyl-CoA Δ5 desaturase as defined herein. The degree of identity in amino acid sequences when the proteins were compared over the entire sequences were: between AL21 and Tribdesat2b, 25%; AL21 and Tribdesat2b, 27%; ALlO and Tribdesat2b, 24%; ALlO and Trib2c, 28%. Therefore, it was concluded that there was insignificant or little homology between the plant acyl-CoA dependent desaturases and the insect ones.
Tribdesatδ and 6a showed activity in yeast cells as Δ9 desaturases that acted primarily and very efficiently on stearic acid to produce oleic acid. They also had some activity on palmitic acid to produce palmitoleic acid. The enzymes did not detectably desaturate substrates of C 14 or shorter when fed to the yeast cells, including C14:0 or C14:1Δ9, or the substrates of C20 or longer including C20:0. They were therefore characterised as stearoyl-CoA Δ9 desaturases.
Tribdesat8 was able to desaturate palmitic acid to produce palmitoleic acid, so it was considered to be a Δ9 desaturase. It did not have detectable desaturase activity in the yeast cells on C 14 or shorter substrates, or on C 18:0 or longer saturated substrates, or on monounsaturated or polyunsaturated substrates including C16:1Δ9, C18:l , C18:2 or C18:3. It therefore appeared to be specific for palmitic acid. The protein had a predicted size of 374 amino acids and included the 8 conserved desaturase motifs and three His boxes (Table 3). On the basis of the sequence homology with known acyl-CoA desaturases, this enzyme was presumed to be acting on the acyl-CoA substrate. This enzyme was therefore characterised as a palmitoyl- CoA Δ9 desaturase.
TribdesatlO showed Δ12 desaturase activity on palmitoleic and oleic acids resulting in C16:2Δ9> Δ12 and C18:2Δ9> Δ12 (LA), respectively. Conversion of oleic acid substrate (at least 30% converted) was about twice as efficient in yeast cells compared to conversion of palmitoleic acid. There was no detectable activity on Cl 6:0, C 18:0 or longer saturated substrates, or Cl 4 or shorter substrates including C14:1Λ9, so the enzyme appeared to be specific as a Δ12 desaturase acting on Δ9-mono-unsaturated substrates of Cl 8 and Cl 6 length joined to CoA. The protein had a predicted size of 366 amino acids and included the 8 conserved desaturase motifs and three His boxes (Table 3). On the basis of the sequence homology with known acyl-CoA desaturases, this enzyme was presumed to be acting on acyl-CoA substrates. This enzyme was therefore characterised as an oleoyl-CoA Δ12 desaturase. It also showed palmitoleoyl- CoA Δ12 desaturase activity. It was believed this is the first gene to be characterised encoding such an enzyme.
Tribdesatll showed Δ9 desaturase activity on saturated substrates having a chain length from C14:0 to C24:0, with the greatest efficiency of conversion observed for C22:0 (known as behenic or docosanoic acid) and C24:0 (known as lignoceric or teracosanoic acid) . Therefore, this enzyme was considered to be a lignoceroyl-CoA Δ9 desaturase, although it also has behenoyl-CoA Δ9 desaturase activity.
CLl showed Δ12 desaturase activity on palmitoleic and oleic acids resulting in C16:2 Λ9' Δ12 and LA, respectively, the same spectrum of activity as TribdesatlO although the activity was less efficient than for TribdesatlO.
CL3 showed efficient desaturation activity on C 14:0 to produce C14:1Δ9. The enzyme did not have detectable activity for C12:0 or C 16:0 and therefore appeared to be a specific myristoyl-CoA Δ9 desaturase. It did not have activity on mono- unsaturated substrates such as C12:1Δ5 or C20:l. On the basis of the sequence homology with known acyl-CoA desaturases, this enzyme was presumed to be acting on the acyl-CoA substrate. This enzyme was therefore characterised as a myristoyl- CoA Δ9 desaturase.
Figure imgf000066_0001
Example 9. Cloning of desaturase genes into Gateway entry vector pENTRl 1.
To construct the functional genes or genes to be tested in a Gateway entry vector, the pGem-T-Easy constructs containing full-length gene sequences were digested with restriction enzymes to excise the inserts. These were separated on a 1% agarose gel and the DNA of the approximately lkb fragments purified using the QIAquick Gel Extraction kit (Qiagen) according to the manufacturer's instructions. The Gateway entry vector pENTR 11 (Invitrogen) was prepared and digested with the required restriction enzymes and dephosphorylated using Calf Intestine Alkaline Phosphatase (Roche). Gene fragments were ligated with the vector and transformed into E.coli strain JM109. Transformants were selected on LB agar plates containing 50μg/ml kanamycin. Clones containing inserts were confirmed by restriction enzyme and sequence analysis.
Cloning of desaturase genes into Gateway destination vectors to create expression clones. Two expression system, SF9 insect cells and Arabidopsis, were selected and
Gateway expression vectors chosen for each. For SF9 insect cells, pDEST8 was chosen and selection for DHlOBac transformants carried out in LB supplemented with kanamycin (50μg/ml), gentamicin (7μg/ml) and tetracycline (lOμg/ml), Bluo-gal (lOOμg/ml) and IPTG (40μg/ml). For Arabidopsis transformation, binary vector pXZP391 (CSIRO Plant Industry) was chosen and selection for transformants carried out in LB supplemented with spectinomycin (50μg/ml). pENTRl 1 clones containing the gene inserts and appropriate pDEST vectors were prepared and under-went an LR recombination reaction according to manufacturers instructions (Gateway LR clonase enzyme mix, Invitrogen). The resultant recombination mixtures were transformed into E.coli strain JM 109 for pXZP391 or DHlOBac (Invitrogen) for pDESTδ. Transformants containing correct inserts were confirmed by restriction enzyme analysis and sequence analysis using vector specific primers.
Example 10. Recombinant Baculovirus production of insect desaturases or acetylenases.
The Tribdesat2b and CL3 genes were cloned into the pDestδ vector
(Invitrogen), using Gateway recombination as above. Competent DHlOBac cells
(Invitrogen) were transformed with the pDestδ clones by heat shock method. Transformants were selected on LB agar containing kanamycin, tetracycline and gentamycin (50μg/mL, lOμg/mL and 7μg/mL respectively) and IPTG and Xgal at the appropriate concentrations, and purified by re-streaking on this medium. Once the recombination events had taken place in DHlOBac, the recombinant bacmids (now called pFastbac) were identified using M 13 forward and reverse primers to colony screen the 2b and CL2 inserts in pFastbac in DHlOBac cells. Positive clones produced a band of approximately 3kb. Clones were picked for downstream applications that did not have any evidence of empty vector contamination which could be observed as a 300bp product.
Transformants containing pFastbac plasmids having the 2b and CL2 inserts were grown overnight in LB medium containing the antibiotics. A modified alkaline lysis plasmid isolation protocol was used according to the manufacturer's instructions in the Bac-to-Bac Expression System manual. Isolated DNA was dissolved in 40μL of IX TE buffer pH 8.0 and quantitated using the Nanodrop spectrophotometer and stored at 40C. Transfecting Insect Cells
Sf9 iβpodopetera frugiperda ovary cell line) cells were seeded in a 6 well tissue culture plate at a rate of 9X105 cells per well, in 2mL of SF900II serum free medium (Invitrogen). The cells were allowed to adhere to the well surface for 2 hours at 270C. For each transfection, lμg of purified bacmid DNA was diluted in lOOuL of unsupplemented Grace's Medium (Invitrogen), in polystyrene tubes. 6μL of Cellfectin® Reagent (Invitrogen) was diluted in lOOμL of unsupplemented Grace's Medium in polystyrene tubes. The DNA and Cellfectin® Reagent were then mixed together and incubated at RT for 30 minutes. While DNA:lipid complexes were incubating, the cells were washed once with 2mL of unsupplemented Grace's Medium and the medium was then removed. 0.8mL of unsupplemented Grace's Medium was added to the DNA.iipid complex, mixed and then added to the washed SF9 cells. The transfection was allowed to proceed at 270C for 5 hours. The DNA:lipid complexes were removed from the cells and 2mL of SF900II were added. Cells were incubated at 270C until signs of infection were obvious. Once the cells appeared infected, the virus was harvested by centrifuging the cell culture medium at lKrpm for 5 minutes at 2O0C. The clarified cell culture medium was transferred to tubes and stored at 40C. This was called Pl viral stock.
Amplifying the baculovirus stocks
Sf9 cells were used to seed 25cm2 flasks at a rate of lX10°cells/mL in 5mL of medium. The cells were infected with Pl viral stock at a multiplicity of infection (MOI) of 0.1 virus particles per cell, which was estimated to be 5X106cells/mL. After 48 hours at 270C, the cells were showing signs of infection and the cell culture medium was harvested as outlined above. This was called P2 viral stock. The P2 stock was then used to produce a P3 stock. 75cm2 flasks were seeded at a rate of lX106cells/mL in 15 mL SF900II medium, and infected at a MOI of 0.1 virus particles per cell, which was estimated to be 5X106cells/mL. After 48 hours at 270C, the cells were showing signs of infection and the cell culture medium was harvested as outlined above. This was called P3 viral stock.
To determine the exact titre of each P3 stock, a TCID50 (Tissue Culture Infectious dose) was performed. A 96 well tray was seeded with 3X103 cells per well. Cells were incubated at 270C overnight. 10-fold dilutions of the virus were prepared to 10"8. 50μL of dilutions 10"3 - 10"8 were added to 8 wells each. The TCID50 was then incubated for 7 days at 270C. The wells were scored positive or negative for virus infection. Once the viruses were titrated, infections of SF9 cells with known multiplicity of infections (MOI) could proceed. Infested cells were removed and dried under vacuum.
Methyl esters of fatty acids of insect cells infected with the construct expressing Tribdesat2b were obtained using the direct acidic methanol reagent as previously described and analysed by GC/MS. The GC/MS trace and spectrum showed C16: 1Δ5 and C18:1Δ5 production by SF9 expressing Tribdesat2b, as compared to the SF9 control. The efficiency of conversion of C16:0 to C16: ld5 in the insect cells transduced with the construct was 16.9% and the efficiency of conversion of C18:0 to C18:ld5 was 29.7%. These data therefore confirmed the data obtained from yeast cells expressing Tribdesat2b and it was concluded that this gene encoded a Δ5-desaturase active on C 16:0 and C 18:0 substrates, with greater activity on the latter. It therefore almost certainly encoded a Stearoyl-CoA Δ5-desaturase, which activity has not previously been reported for a cloned gene. The other desaturases are likewise expected to show activity in insect cells.
Example 1 1. Expression of Tribolium and Chauliopnathus genes in plants
The Tribolium TribdesatlO and 2b genes were cloned into pENTRl l as described above and then recombined into plant expression vector pXZP391 which provides for expression of the coding region insert under the control of a seed-specific napin promoter (FpI) isolated from Brassica napus (Stalberg et al., 1993). The expression plasmid was transformed into Agrobacterium tumefaciens AGLl, and used for plant transformation into the Arabidopsis thaliana fad2/fael double mutant which lacks Δ12-desaturase activity, by in planta method. Seeds of spray-inoculated plants were harvested 3-4 weeks later and plated onto selective media. Tl transformed plants were identified on MS media containing 40mg/L kanamycin and 100mg/L timentin, and transferred into pots in the glasshouse. T2 seeds from each Tl plant were analysed for fatty acid composition by gas chromatography. The data shown in Figure 6 demonstrated the Δ12 desaturase activity of TribdesatlO in Arabidopsis, as LA was produced from oleic acid in the fad2/fael mutant plant.
Expression of transgenes in tobacco colli and plants
The Tribdesat 2b, CLl and CL2 coding regions were cloned in the 'sense' orientation into the plant expression vector pVEC8 (Wang et al., 1997) under the control of the regulatory sequences CaMV 35S promoter and octopine synthase transcription termination/polyadenylation signal (Gleave, 1992), providing strong constitutive expression of the transgene in most plant tissues. The transgene was introduced into tobacco by Agrobacterium-mediaXed transformation of leaf tissue (Horsch et al., 1985) with selection of the transgenic cell lines on hygromycin- containing media. Transgenic plant cells were selected based on the resistance to hygromycin, as conferred by a hpt resistance marker gene in the pVEC8 derived vector. Transformed tobacco lines were cultured to produce either calli or regenerated to produce whole plants, depending on the phytohormone regime in the selection and growth media (Murashige and Skoog, 1962; Horsch et al., 1985). Undifferentiated transgenic tobacco calli were produced by selection on calli-inducing phytohormones (0.5 mg/L indole acetic acid (IAA) and 0.05 mg/L kinetin) and calli were maintained on agar plates. Differentiated tobacco plants were produced by first selecting transgenic material on shoot-inducing media by the addition of shoot-inducing phytohormones (100 μg/L N-6-benzyladinine (BAP) and 500 ng/L L IAA). After 3 weeks of growth on this media individual tobacco apices were cut from the leaf discs and re-plated onto root-inducing media containing root-inducing phytohormones (50 ng/L IAA). A further 3 weeks are required for the formation of roots. Transgenic material, either calli or whole plants, were selected for analysis as required. The Tribdesat2b gene was also recombined using LR clonase into plant expression vector pXZP393 (CSIRO Plant Industry) under control of the constitutive CaMV 35S promoter. The resulting expression plasmid pXZP384 was transformed into AGLl, and used for plant transformation into tobacco leaves as described above. Fatty acid composition of transgenic tobacco leaves and seed will be analysed by GC and GC-MS to shown modification of fatty acid composition by addition of the new desaturase activity. Example 12. Cloning of desaturase genes from Acheta domesticus.
Some insects are thought to possess Δ12-desaturase activity but as yet no gene encoding such an enzyme has been isolated despite extensive effort over 20 years. The reasons for this failure were unknown until the present work. One insect known to possess Δ12-desaturase activity is Acheta domesticus, the common cricket. In an initial attempt to clone an A. domesticus Δ12-desaturase gene, a set of degenerate primers AdD12Des-Fl (5'-TTGTTCTGTGTGGGTCAYGAYTGYGGWCA (SEQ ID NO: 127)) and AdD12Des-Rl (5'-GTGATGGGCGACGTGACYGTYKGTRAT (SEQ ID NO: 128) were designed based on the conserved histidine Box I and histidine Box III regions of A. thaliana Δ12-desaturase FAD2 (P46313), Caenorhabditis elegans Δ12- desaturase FAT-2 (AAF63745) and Δ15-desaturase FAT-I (L41807), corresponding to LFCVCHDCGH (amino acid residues 88-97) and ITNGHVAHH (amino acid residues 291-299) of FAT-2. However, RT-PCR reactions using A. domesticus fat body total RNA with this set of primers failed to amplify any specific product despite repeated attempts and varied conditions, suggesting that the A. domesticus gene(s) encoding Δ12-desaturase might share low homology to the plant or nematode Δ12-desaturase genes.
A different approach was considered to clone the A. domesticus Δ12-desaturase gene. Based on the amino acid sequence homology shared among 55 insect fatty acid acyl-CoA Δ9-, Δ10-, Δl l- and Δ14-desaturases from species including Argyrotaenia velutinana (AAF44709), Bombyx mori (BAD18122, AAF80355), Epiphyas postvittana (AAK94070), Helicoverpa assult (AAM28484, AAM28483), H. zea (AAF81787, AAF81788), Musca domestica (AAN31393), Ostrinia furnacalis (AAL27034, AAL32060, AAL35746), O. nubilalis (AAF44710, AAL35330, AAL35331), Trichoplusia ni (AAB92583, 044390, AAD03775) etc, degenerate primers (AdD12Des-F2 5'- TTCTTCTTCKCNCAYKTHGGNTGG (SEQ ID NO: 129) and AdD12Des-R2 5 '-TGRTGGTAGTTGTGVHANCCCTC (SEQ ID NO: 130)) were designed that targeted two conserved regions of these desaturases (FFFS/ AHI/VGW (SEQ ID NO: 131) and EGY/W/FHNYHH (SEQ ID NO: 132), corresponding to amino acid residues 145-152 and 263-270 of A. velutinana Δ9-desaturase AAF44709) in an attempt to clone an insect Δ12-desaturase gene. Surprisingly, RT-PCR from A. domesticus fat body total RNA amplified a divergent 360 bp desaturase-like sequence, along with another desaturase gene fragment identical to part of the known A. domesticus Δ9-desaturase (AdD9Des, AF338466). Rapid amplification of cDNA ends using GeneRacer Kit (Invitrogen) and sequences internal to the 360bp fragment resulted in a 1597 bp cDNA sequence that was designated AdDl 2Des (SEQ ID NO: 133) coding for a peptide of 357 amino acid residues (SEQ ID NO: 134).
The cDNA sequences of AdD9Des, AdD12Des and TribdesatlO were each cloned in yeast expression vector pYES2 (Invitrogen), generating plasmids pXZP277, pXZP282 and pYES2-TriblO respectively. Plasmids pYES2, pXZP277, pXZP282 and pYES2-TriblO were transformed into Saccharomyces cerevisiae cell of the strain olel which contains a Δ9-desaturase knock-out mutation, (Stukey et al., 1990) in order to test for complementation of this mutation and therefore indicate Δ9-desaturase activity. Yeast olel cells needed to be supplied with an unsaturated fatty acid such as C 16:1 Λ9 to maintain growth, unless a Δ9-desaturase is expressed within the cells, which was the basis of the complementation test. Transformants in olel cells were selected on dropout media (SD-Ura) agar plate supplied with 0.5 mM C16:1Δ9 and 1% NP-40. Yeast olel transformants carrying the above mentioned plasmids were first grown in YPD media (1% yeast extract, 2% peptone, 2% dextrose), supplied with 0.5 mM C16:1Δ9 and 1% NP-40 at 3O0C until OD600 around 1.0. The samples were then adjusted to an OD600 of 1.0, diluted in 1:10 series and 1 μL aliquots of each dilution spotted onto YPG agar plates, which was the same as YPD media except that it contained 2% galactose instead of dextrose to induce the gene expression from the pYES2 derived vectors. The growth of cells was observed after 2 days at 3O0C. Expression of A. domesticus Δ9-desaturase in olel cells clearly complemented the olel phenotype, while expression of A. domesticus AdDl 2Des did not complement, indicating that it was not functioning as a Δ9-desaturase in yeast cells. The olel cells expressing the T. castaneum Δ12-desaturase were shown to grow on YPG plate only in the presence of added C 17:1. These data established that the cloned AdDl 2Des or TribdesatlO genes did not encode another member of a Δ9-desaturase family.
These expression plasmids were also transformed into Saccharomyces cerevisiae S288C cells, which is a common laboratory yeast strain. Fatty acid methyl esters (FAME) extraction from yeast cells and analysis were performed as described by Zhou, et al. (2006). Expression oϊAdD12Des in S. cerevisiae produced new diene fatty acids C 18:2 and C 16:2 from C 18:1 and C 16:1, respectively, as demonstrated by gas chromatography (GC) of yeast fatty acid methyl esters (Fig. 7A) when compared to cells with pYES2 vector only (Fig. 7B), confirmed by the identical retention time and mass spectrum as the pure C18:2Δ9>12 standard in the first instance. The double bond positions of the C 16:2 and C 18:2 products were confirmed by GC-mass spectrometry of the 4,4-dimethyloxazoline derivative (Fay and Richli, 1991) to be at the Δ9 and Δ 12 positions by the gap of 12 atomic mass units between m/z 196 and 208, 236 and 248 (Fig. 2C). The production of these dienes demonstrated that AdDHDes and TribdesatlO encoded Δ12-desaturases acting on C16:1Λ9 or C18:1A9.
When expressing in cells of yeast strain olel, T. castaneum Δ12-desaturase produced C18:2 from C18:l at conversion rate of 30.5%, and C16:2 from C16:l at the conversion rate of 16.0% (Table 11), indicating a preference of the enzyme for the Cl 8 substrate over the C16 substrate. There was a concomitant decrease in the level of substrate fatty acid in the cells.
Table 11. Fatty acid composition (% total fatty acids) of yeast olel cells expressing Tribolium castaneum desaturase TribdesatlO in pYES2 when fed C16:l or C18: l substrates, compared to an empty pYES2 vector control.
Figure imgf000073_0001
The A. domes ticus and T. castaneum Δ12-desaturase protein coding regions were also cloned into a plant expression vector to generate plasmid pXZP375 and pXZP376 respectively, under control of the seed specific promoter FpI (Stalberg et al.,
1993). These expression plasmids were introduced into tissues of an A. thaliana fad2/fael double mutant (Smith et al., 2003) to produce transformed plants via
Agrobacterium tumefaciens as described by Zhou et al., (2006). GC analysis of seed FAMEs was essentially as described above. Shown in Table 12 as an example, six transgenic lines transformed with the AdDl 2Des gene encoding the A. domesticus Δ12- desaturase efficiently produced C 18:2, with levels obtained of at least 42% of the total fatty acid in the seedoil, and at least 20% C18:3, both of which were produced in the seed from Cl 8:1. Since the A. thaliana fad2/fael double mutant still had the wild-type Δ15-desaturase (FAD3), this enzyme was presumed to desaturate C18:2, when available, into C18:3. Therefore, the C18:3 produced in these transgenic lines was also a product of the conversion of C 18:1 to C 18:2 by the A. domes ticus Δ12-desaturase, and represented A. domes ticus Δ12-desaturase product. This result confirmed the Δ12- desaturase activity of the cloned gene, and represented the first demonstration of a recombinant insect Δ12-desaturase in transformed cells.
Table 12. Fatty acid composition (% total fatty acids) of transgenic Arabidopsis seeds expressing A domes ticus AdD12Des in fad2/fael double mutant.
Figure imgf000074_0001
* fad2/fael corresponds to the control (untransformed) plants
Phylogenetic analysis of representative acyl-CoA or acyl-lipid desaturase protein sequences showed that the amino acid sequences of AdD12Des and TribdesatlO clustered with other acyl-CoA desaturases (right hand side of Figure 9; Figure 10). The accession numbers and species for insect desaturases used in the analysis were: AdD9, Acheta domesticus AAK25796; AdD 12, A. domesticus SEQ ID NO: (SEQ ID NO: 135); BmDlO-I l, Bombyx mori AAF80355; BmDI l, B. mori BAD18122; DmD9, Drosophila melanogaster CAB69054; MdD9, Musca domesticus AAN31393; OnD9, Ostrinia nubilalis AAF44710; OnDI l, O. nubilalis AAL35331; OnD14, O. nubilalis AAL35330; PoD9, Planotortrix octo AAF73073; PoDlO, P. octo AAG54077; TcD9A, Tribolium castaneum XP 969607; TcD9B, T. castaneum XP 966962; TcD 12, T. castaneum TribdesatlO (SEQ ID NO: 28); TnD9, Trichoplusia ni AAB92583; TnDI l, T. ni 044390; other animal desaturases AcD12-15, Acanthamoeba castellanii ABK15557; CeD12, Caenorhabditis elegans AAF63745; RnD9, Rattus norvegicus P07308; bacterial desaturases N36D9, Nostoc sp. 36 CAF18423ΔNoD9, Nostoc sp. PCC 7120 (Anabaena variabilis) BAA03434; NoD9, Nostoc sp. PCC 7120 BAA03435; SyD9, Synechocystis PC 6803 BAA03982; SyD12, Synechocystis PC 6803 P20388; fungal desaturases LkD12, Lachancea kluyveri BAD08375; MaD9, Mortierella alpina BAA75927; Y1D12, Yarrowia lipolytica CAJ81209; ScD9, Saccharomyces cerevisiae P21147; plant desaturases AtD9, Arabidopsis thaliana AAK76592; AtD12_FAD2 (ER Δ12-desaturase), Arabidopsis thaliana P46313; AtD12_FAD6 (plastid Δ12-desaturase), A. thaliana P46312; PgD9, Picea glauca AAM12238). D9, DlO, DI l, D12, and D14 designate Δ9-, Δ10-, Δ11-, Δ12-, and Δ14- desaturases, respectively.
AdD12Des and TribdesatlO showed only low identities (up to 23%) over the full-length amino acid sequences to bacterial Δ9-desarurases or plant, fungal and animal Δ12-desaturases which formed another major cluster of predominantly acyl- lipid desaturases (left hand side of Figure 4). The low level of identity of AdD12Des and TribdesatlO with Δ12-desaturases from other animals, plants or fungi was particularly unexpected. Instead, AdD12Des formed a discrete cluster with insect Δ9- desaturases including those of house cricket and red flour beetle suggesting its possible evolution from Δ9-desaturase genes. The extent of identity in amino acid sequence of AdD12Des to these were 65.2%, 55.2%, 55.5%, 53.5%, 53.5%, 55.3%, 59.4% and 53.6%, respectively, to insect Δ9-desaturases from A. domesticus AF338466, D. melanogaster CAB69054, M. domesticus AAN31393, O. nubilalis AAF44710, P. octo AAF73073, T. castaneum XP_969607, T. castaneum XP_966962 and T. ni AAB9258. In particularly, AdD12Des was relatively close in sequence to the Δ9-desaturase protein from the same species, sharing about 65% amino acid identity.
As TribdesatlO appeared on a separate branch of the phylogenic tree to the other acyl CoA desaturases including Tribolium Δ9-desaturases to which it shared up to 48% identity, it may have evolved independently of the insect Δ9-desaturase genes. As shown by the data in Figure 3, AdD12Des retained low levels of Δ9-desaturation activity on the medium chain saturated fatty acids, C 14:0 and Cl 5:0, when expressed in yeast. Figure 3A shows the partial GC graph of fatty acid profile in yeast cells expressing pYES2 vector only, while 3B shows fatty acid profile in yeast cells expressing pXZP282, with extra C 14:1 and C 15:1 peaks. However, no detectable Δ9- desaturation activity was seen in yeast expressing TribdesatlO. These findings suggest that these two insect Δ12-desaturases independently diverged from an ancient desaturase. It was also noteworthy that the other animal Δ12-desaturases from Caenorhabditis elegans and Acanthamoeba castellanii were more closely related to the plant Δ12-desaturases than to insect Δ12-desaturases AdD12Des and TribdesatlO (27.2% and 38.6% amino acid sequence identities to Arabidopsis thaliana Δ12- desaturase FAD2, respectively, or 11.6% and 11.1% amino acid sequence identities to A. domesticus AdD12Des, respectively). To test whether the cloned A. domesticus Δ12-desaturase used acyl-CoA Cl 8:1 as substrate rather than acyl-PC Cl 8:1, the Δ12-desaturase gene from A. domesticus in pXZP282 was expressed in yeast S288C cells. The Arabidopsis thaliana Δ12- desaturase gene coding region encoding FAD2 was also cloned into pYES2, resulted in the construct pXZP279, and expressed in S288C as a control gene, as an example of a plant Δ12-desaturase that was known to be an acyl-PC type enzyme. Cultures of the yeast transformants were incubated in the presence of C14 labelled C 18 : 1 (5xlO6 dpm) and cells were harvested at 2, 5, and 30 minutes, and 1, 6 and 24 hours later. For optimal results, cells were lysed using glass beads followed by repeated passes through a tissue grinder. Fatty acids bound to CoA were extracted as described by Domergue et al. (2005), converted to FAMEs and separated by argentation-thin layer chromatography (TLC). Oleic acid added to the medium during induction became immediately available in the acyl-CoA, PC and TAG fractions. It was observed that Arabidopsis FAD2 accumulated labelled C 18:2 product in the acyl-CoA fraction from 30 minutes onwards. In contrast, conversion of labelled C18:1 to C18:2 by the cricket desaturase was observed as early as 2 minutes after addition of the substrate, indicating that this enzyme uses acyl-CoA as substrate. Therefore, AdD12Des, and presumably TribdesatlO, were of the acyl-CoA oleoyl Δ12-desaturase type, rather than acyl-PC oleoyl Δ12-desaturases. Genes encoding the former class of Δ12-desaturase have not previously been identified.
Cloning of the insect acyl-CoA Δ12-desaturases described herein is expected to stimulate the discovery of other novel insect polyunsaturated fatty acid desaturase genes and provide greater insight into insect fatty acid metabolism. The role of this gene in insect development and physiology can be studied in knockout mutants. Characterization of these Δ12-desaturase genes will also shed more light on the evolution of insect desaturases via ancestral genes. Finally, this will increase our understanding of sequence-substrate type relationships among fatty acid desaturases, since membrane-associated insect Δ12-desaturases are thought to use acyl-CoA substrates (Cripps, et al. 1990), while membrane-associated plant Δ12-desaturases utilize phosphatidylcholine (PC) as a substrate (Stymne and Appelqvist, 1978). REFERENCES
Abdullah et al. (1986) Biotechnology 4:1087.
Altschul et al. (1997) Nucleic Acids Res 25: 3389-3402. Badami and Patil (1981) Prog. Lipid Res 19: 119-153. Baumlein et al. (1991) MoI. Gen. Genet. 225:459-467. Baumlein et al. (1992) Plant J. 2:233-239. Broun et al. (1998) Plant J. 13:201-210. Cahoon et al. (2001) J Biol Chem 276:2637-2643. Cahoon, et al. (2000) Plant Physiology 124, 243-251. Cahoon, et al (1992) Proc Nat Acad Sci USA 89: 11184-11 188. Capecchi (1980) Cell 22:479-488. Cheng et al. (1996) Plant Cell Rep. 15:653-657.
Christie (2003) Lipid Analysis: isolation, separation, identification and structural analysis of lipids. 3rd edition, The Oily Press, Bridgwater, UK. Clapp (1993) Clin. Perinatol. 20:155-168.
Cripps et al. (1990) Arch Biochem Biophys, 278, 46. Crombie et al. (1984) J Chem Soc Chem Commun 15:953-955. Crombie et al. (1985) J Chem Soc Perkin Trans 1:2425-2434.
Cuperus and Derksen (1996) High Value- Added Applications from Vernolic Acid. p. 354-356. In: J. Janick (ed.), Progress in new crops. ASHS Press, Alexandria, VA.
Curiel et al. (1992) Hum. Gen. Ther. 3:147-154.
Domergue et al. (2005) Biochem J. 389, 483-490.
De Renobales et al. (1987) Trends in Biochemical Sciences 12, 364.
Dobson and Christie (2002) European Journal of Lipid Science and Technology, 104: 36-43.
Eglitis et al. (1988) Biotechniques 6:608-614. Fay and Richli (1991) J. Chromatography 541, 89-98. Fritsche et al. (1999) FEBS Letters 462:249-253. Fujimura et al. (1985) Plant Tissue Culture Letters 2:74. Gleave (1992). Plant Molecular Biology 20, 1203-1207. Graham et al. (1973) Virology 54:536-539. Grant et al. (1995) Plant Cell Rep. 15:254-258.
Gunstone et al. (1994) (Editors), The Lipid Handbook (2nd Edition). Chapman & Hall, London).
Hao et al. (2002) Insect Biochem MoI Biol 32 961-966. Harayama (1998). Trends Biotechnol 16: 76-82.
Horsch et al. (1985). Science 227, 1229-1231.
Kleiman and Spencer (1982) J Am Oil Chem Soc 59, 29-38.
Knipple et al. (2002) Genetics 162: 1737-1752.
Koziel et al. (1996) Plant MoI. Biol. 32:393-405. Lee et al. (1998) Science 280:915-8.
Lewis et al. (2000) J Microbiol Methods 43: 107-116.
Lu et al. (1993) J. Exp. Med. 178:2089-2096.
Mortimer and Johnston (1986) Genetics 113: 35-43.
Moto et al. (2004) Proc Natl Acad Sci USA 101: 8631-8636. Murashige and Skoog (1962). Physiologia Plantarum 15, 473-477.
Murata and Wada (1995) Biochem. J. 308:1-8.
Needleman and Wunsch (1970) J. MoI. Biol. 48:443-453.
Qiu et al. (2001) Plant Physiol 125:847-855.
Rodriguez et al. (2004) Insect Biochem MoI Biol 34: 1315-1328. Sayanova et al (2007) Plant Physiology 144: 455-467.
Serra et al. (2006) Insect Biochem MoI Biol 36:822-825.
Shanklin and Cahoon (1998) Annu. Rev. Plant Physiol. Plant MoI. Biol. 49:611-641.
Simopoulos (2000) Poultry Science 79:961-970.
Smith (1971) Prog. Chem. Fats other Lipids 11:137-177. Smith et al. 2003. Planta 217, 507-516.
Stalberg et al.(1993) Plant Molecular Biology 23, 671-683.
Stukey et al. 1990. J Biol Chem 265, 20144-20149.
Stymne and Appelqvist (1978). Eur J Biochem 90, 223-229.
Thompson et al. (1997) Nucleic Acids Res. 25: 4876-4882. Toriyama et al. (1986) Theor. Appl. Genet. 205:34.
Trautwein (2001) Eur. J. Lipid Sci. Technol. 103:45-55. van de Loo et al. (1995) Proc Natl Acad Sci U S A. 92:6743-7. Vernet et al. (1987) Gene, 52: 225-233.
Wagner et al. (1992) Proc. Natl. Acad. Sci. USA 89:6099-6103. Wang et al. (1997) The Journal of Genetics & Breeding 51 : 325-334. Whittle et al. (2005) J Biol Chem. 280:28169-76. Yu et al. (1988) Lipids 23: 804-810. Zhou et al. (2006) Plant Science 170, 665-673.

Claims

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
1. A eukaryotic cell comprising an exogenous nucleic acid encoding a polypeptide which is: (i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs :16 to 30, 73 to 78, 80 and 134,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and/or 134, and/or (iii) a biologically active fragment of i) or ii), wherein the polypeptide has one or more activities selected from desaturase, conjugase, epoxidase and hydroxylase activity.
2. The eukaryotic cell of claim 1, wherein the polypeptide comprises amino acids having a sequence which is at least 90% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and 134.
3. The eukaryotic cell of claim 1 or claim 2, wherein the polypeptide can be isolated from an insect of the Order Coleoptera or Orthoptera.
4. The eukaryotic cell of claim 3, wherein the insect is a species of Tribolium, Chauliognathus or Acheta.
5. The eukaryotic cell of any one of claims 1 to 4, wherein the polypeptide is: (i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs: 28, 73, and/or 134,
(ii) a polypeptide comprising amino acids having a sequence which is at least
50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134, and/or (iii) a biologically active fragment of (i) or (ii), wherein the polypeptide has acyl-CoA Δ12 desaturase activity.
6. The eukaryotic cell of any one of claims 1 to 4, wherein the polypeptide is:
(i) a polypeptide comprising amino acids having a sequence as set forth in SEQ ID NO: 18 or SEQ ID NO: 19, (ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to a sequence set forth in SEQ ID NO: 18 or SEQ ID NO: 19, and/or
(iii) a biologically active fragment of (i) or (ii), wherein the polypeptide has acyl-CoA Δ5 desaturase activity.
7. The eukaryotic cell of any one of claims 1 to 4, wherein the polypeptide is:
(i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs: 17, 22, 23, 27, 29, 74, 75 and/or 76,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 17, 22, 23, 27, 29, 74, 75 and/or 76, and/or
(iii) a biologically active fragment of (i) or (ii), wherein the polypeptide has acyl-CoA Δ9 desaturase activity.
8. A eukaryotic cell comprising an exogenous nucleic acid encoding an acyl-CoA Δ12 desaturase.
9. The eukaryotic cell of claim 8, wherein the acyl-CoA Δ12 desaturase comprises: (i) amino acids having a sequence as set forth in any one of SEQ ID NOs: 28, 73, and/or 134,
(ii) amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134, and/or (iii) a biologically active fragment of (i) or (ii).
10. The eukaryotic cell of claim 8 or claim 9 which comprises an increased level of 16:2Δ9'Δ12 and/or 18:2Δ9>Δ12 fatty acids relative to a corresponding eukaryotic cell lacking the exogenous nucleic acid.
11. The eukaryotic cell according to any one of claims 8 to 10 which comprises an increased level of 16:2Δ9>Δ12 and/or 18 :2Δ9>Δ12 fatty acids which are esterified to CoA.
12. A eukaryotic cell comprising an exogenous nucleic acid encoding an acyl-CoA Δ5 desaturase, wherein the desaturase is more active on a 18:0 and/or 16:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is any one, two, three or all of a 18:1Δ9, 16:1Δ9, 20:0 and 20:2Δl lΔI4.
13. The eukaryotic cell of claim 12, wherein the acyl-CoA Δ5 desaturase comprises: (i) amino acids having a sequence as set forth in SEQ ID NO: 18 or SEQ ID
NO: 19,
(ii) amino acids having a sequence which is at least 50% identical to SEQ ID NO: 18 or SEQ ID NO: 19, and/or
(iii) a biologically active fragment of (i) or (ii).
14. The eukaryotic cell of claim 12 or claim 13 which comprises an increased level of 16: 1Δ5 and/or 18 : 1 Δ5 fatty acids relative to a corresponding eukaryotic cell lacking the exogenous nucleic acid.
15. The eukaryotic cell according to any one of claims 12 to 14 which comprises an increased level of 16:1A5 and/orl8:lΔ5 fatty acids which are esterified to CoA.
16. A eukaryotic cell comprising an exogenous nucleic acid encoding an acyl-CoA Δ9 desaturase, wherein the desaturase is more active on a 14:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is 16:0 and/or 18:0.
17. The eukaryotic cell of claim 16, wherein the acyl-CoA Δ9 desaturase comprises: (i) amino acids having a sequence as set forth in SEQ ID NO: 23 or SEQ ID
NO: 74,
(ii) amino acids having a sequence which is at least 50% identical to SEQ ID NO: 23 or SEQ ID NO:74, and/or
(iii) a biologically active fragment of (i) or (ii).
18. The eukaryotic cell of claim 16 or claim 17 which comprises an increased level of 14: 1Δ9 relative to a corresponding eukaryotic cell lacking the exogenous nucleic acid.
19. The eukaryotic cell according to any one of claims 16 to 18 which comprises an increased level of 14: 1Δ9 which is esterified to CoA.
20. The eukaryotic cell according to any one of claims 1 to 19 which is a plant cell, a mammalian cell, an insect cell, a fungal cell or a yeast cell.
21. The eukaryotic cell of claim 20 which is in a plant or a plant seed.
22. The eukaryotic cell of claim 21, wherein the plant or plant seed is an oilseed plant or an oilseed respectively.
23. A process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
(i) obtaining a nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence more closely related to SEQ ID NO: 28 than to SEQ ID NO: 135,
(ii) introducing the nucleic acid molecule into a cell or cell-free expression system in which the promoter is active,
(iii) determining whether the fatty acid composition is modified relative to the cell or cell-free expression system before introduction of the nucleic acid molecule, and
(iv) optionally, selecting a nucleic acid molecule which modified the fatty acid composition.
24. A process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
(i) obtaining a nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence that is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134,
(ii) introducing the nucleic acid molecule into a cell or cell-free expression system in which the promoter is active,
(iii) determining whether the fatty acid composition is modified relative to the cell or cell-free expression system before introduction of the nucleic acid molecule, and
(iv) optionally, selecting a nucleic acid molecule which modified the fatty acid composition.
25. The process of claim 23 or claim 24, wherein step (iv) comprises selecting a nucleic acid molecule encoding an acyl-CoA Δ12 desaturase.
26. The process according to any one of claims 23 to 25, wherein the modified fatty acid composition comprises an increased level of 16:2Δ9>Δ12 and/orl8:2Δ9'Δ12.
27. A process for identifying a nucleic acid molecule involved in fatty acid modification comprising: (i) obtaining a nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence that is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 18 and/or 19, (ii) introducing the nucleic acid molecule into a cell or cell-free expression system in which the promoter is active,
(iii) determining whether the fatty acid composition is modified relative to the cell or cell-free expression system before introduction of the nucleic acid, and
(iv) optionally, selecting a nucleic acid molecule which modified the fatty acid composition.
28. The process of claim 27, wherein step (iv) comprises selecting a nucleic acid molecule an acyl-CoA Δ5 desaturase, wherein the desaturase is more active on a 18:0 and/or 16:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is any one, two, three or all of a 18:1Δ9, 16:1Δ9, 20:0 and 20:2Δ11Δ14.
29. The process of claim 27 or claim 28, wherein the modified fatty acid composition comprises an increased level of 16:1Δ5 and/or 18 : 1 A5 fatty acids.
30. A process for identifying a nucleic acid molecule involved in fatty acid modification comprising:
(i) obtaining a nucleic acid molecule operably linked to a promoter, the nucleic acid molecule encoding a polypeptide comprising amino acids having a sequence that is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 17, 22, 23, 27, 29, 74, 75 and/or 76,
(ii) introducing the nucleic acid molecule into a cell or cell-free expression system in which the promoter is active,
(iii) determining whether the fatty acid composition is modified relative to the cell or cell-free expression system before introduction of the nucleic acid, and (iv) optionally, selecting a nucleic acid molecule which modified the fatty acid composition.
31. The process of claim 30, wherein step (iv) comprises selecting a nucleic acid molecule encoding an acyl-CoA Δ9 desaturase, wherein the desaturase is more active on a 14:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is 16:0 and/or 18:0.
32. The process of claim 30 or claim 31, wherein the modified fatty acid composition comprises an increased level of 14: 1Δ9.
33. The process according to any one of claims 23 to 32, wherein the polypeptide is an insect polypeptide or mutant thereof.
34. A substantially purified or recombinant polypeptide which is an acyl-CoA Δ12 desaturase.
35. The polypeptide of claim 34, wherein the polypeptide is:
(i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs: 28, 73, and/or 134,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 28, 73, and/or 134, and/or
(iii) a biologically active fragment of (i) or (ii).
36. A substantially purified or recombinant polypeptide which is an acyl-CoA Δ5 desaturase, wherein the desaturase is more active on a 18:0 and/or 16:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is any one, two, three or all of a 18:1Δ9, 16:1Λ9, 20:0 and 20:2Δ11ΔM.
37. The polypeptide of claim 36, wherein the polypeptide is: (i) a polypeptide comprising amino acids having a sequence as set forth in SEQ
ID NO: 18 or SEQ ID NO: 19,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to SEQ ID NO: 18 and/or SEQ ID NO: 19, and/or
(iii) a biologically active fragment of (i) or (ii).
38. A substantially purified or recombinant polypeptide which is an acyl-CoA Δ9 desaturase, wherein the desaturase is more active on a 14:0 substrate than on a fatty acid substrate esterified to CoA, wherein the fatty acid is 16:0 and/or 18:0.
39. The polypeptide of claim 38, wherein the polypeptide is: (i) a polypeptide comprising amino acids having a sequence as set forth in SEQ ID NO: 17 or SEQ ID NO:74,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to one or more of the sequences as set forth in SEQ ID NO: 17 and/or SEQ ID NO: 74, and/or
(iii) a biologically active fragment of (i) or (ii).
40. A substantially purified or recombinant polypeptide which is:
(i) a polypeptide comprising amino acids having a sequence as set forth in any one of SEQ ID NOs :16 to 30, 73 to 78, 80 and 134,
(ii) a polypeptide comprising amino acids having a sequence which is at least 50% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and/or 134, and/or
(iii) a biologically active fragment of i) or ii), wherein the polypeptide has one or more an activities selected from desaturase, conjugase, epoxidase and/or hydroxylase activity.
41. The polypeptide of claim 40, wherein the polypeptide comprises amino acids having a sequence which is at least 90% identical to any one or more of the sequences set forth in SEQ ID NOs: 16 to 30, 73 to 78, 80 and/or 134.
42. The polypeptide according to any one of claims 34 to 41, wherein the polypeptide can be isolated from an insect of the Order Coleoptera or Orthoptera.
43. The polypeptide according to any one of claims 40 to 42, wherein the polypeptide has desaturase activity upon a carbon-carbon bond located at any one of the Δ2 to Δ15 positions of a fatty acid.
44. The polypeptide of claim 43, wherein the fatty acid is a C16 or C18 fatty acid.
45. The polypeptide according to any one of claims 34 to 44, which is a fusion protein further comprising at least one other polypeptide sequence.
46. An isolated and/or exogenous polynucleotide comprising: (i) a sequence of nucleotides selected from any one of SEQ ID NOs: 1 to 15, 67 to 72, 79 and 133, (ii) a sequence of nucleotides encoding a polypeptide according to any one of claims 34 to 45,
(iii) a sequence of nucleotides which are at least 50% identical to one or more of the sequences set forth in SEQ ID NOs: 1 to 15, 67 to 72, 79 and/or 133, and/or (iv) a sequence which hybridises to any one of (i) to (iii) under stringent conditions.
47. A vector comprising the polynucleotide according to claim 46.
48. The vector of claim 47, wherein the polynucleotide is operably linked to a promoter.
49. A cell comprising the recombinant polypeptide according to any one of claims 34 to 45, the exogenous polynucleotide of claim 46 and/or the vector of claim 47 or claim 48.
50. The cell of claim 49 which is a plant, fungal, yeast, bacterial or animal cell.
51. A method of producing the polypeptide according to any one of claims 34 to 45, the method comprising expressing in a cell or cell free expression system the polynucleotide of claim 46.
52. The method of claim 51, further comprising isolating the polypeptide.
53. A transgenic non-human organism comprising a cell according to any one of claims 1 to 22, 49 or 50.
54. The organism of claim 53 which is a transgenic plant.
55. A seed comprising the cell according to any one of claims 1 to 21, 49 or 50.
56. Oil produced by, or obtained from, the cell according to any one of claims 1 to 21, 49 or 50, the transgenic non-human organism of claim 53 or 54, or the seed of claim 55.
57. The oil of claim 56 which comprises fatty acids 16:0, 16:1Δ5, 18:0 and 18:1Δ5, wherein the ratio of the total amount of 18 : 1 A5 to 18:0 in the oil is between 100:1 and 1 :2, and wherein the fatty acid of the oil comprises less than 10% (w/w) 20: 1A5.
58. The oil of claim 56 or claim 57, wherein at least 43% of the C18 fatty acid of the oil is 18:1A5.
59. The oil according to any one of claims 56 to 58, wherein the fatty acid of the oil comprises at least 3.0% (w/w) 18 : 1 A5 as a percentage of the total fatty acid of the oil.
60. The oil according to any one of claims 56 to 59, wherein the oil comprises less than 10% (w/w) 18:3Δ9'Δ12'Δ15 (ALA) as a percentage of the total fatty acid in the oil.
61. The oil according to any one of claims 56 to 60, wherein the oil is obtained by extraction of oil from an oilseed.
62. An oil comprising fatty acids 16:0, 16:1Δ5, 18:0 and 18:1Δ5, wherein the ratio of the total amount of 18:1 Δ5 to 18:0 in the oil is between 100:1 and 1:2, and wherein the fatty acid of the oil comprises less than 10% (w/w) 20: 1Δ5.
63. An oil comprising fatty acids, wherein at least 43% of the Cl 8 fatty acid of the oil is 18:1Δ5.
64. An oil comprising fatty acids comprising at least 3.0% (w/w) 18 : 1 Δ5 as a percentage of the total fatty acid of the oil.
65. The oil of claim 64 which comprises less than 10% (w/w) 18:3A9'A12'Λ15 (ALA) as a percentage of the total fatty acid in the oil.
66. A fatty acid produced by, or obtained from, the cell according to any one of claims 1 to 21, 49 or 50, the transgenic non-human organism of claim 53 or 54, or the seed of claim 55.
67. A method of producing oil containing unsaturated fatty acids, the method comprising extracting oil from the cell according to any one of claims 1 to 21, 49 or 50, the transgenic non-human organism of claim 53 or 54, or the seed of claim 55.
68. A composition comprising a cell according to any one of claims 1 to 21, 49 or 50, the polypeptide according to any one of claims 34 to 45, a polynucleotide according to claim 46, a vector of claim 47 or claim 48, an oil according to any one of claims 56 to 65 or a fatty acid of claim 66.
69. Feedstuffs, cosmetics or chemicals comprising the oil according to any one of claims 56 to 65 or a fatty acid of claim 66.
70. A method of performing a desaturase reaction, the method comprising contacting a substrate saturated, monounsaturated or polyunsaturated fatty acid esterified to CoA with the polypeptide of any one of claims 34 to 45.
71. A substantially purified antibody, or fragment thereof, that specifically binds a polypeptide of any one of claims 34 to 45.
72. A method of treating or preventing a condition which would benefit from a PUFA, the method comprising administering to a subject a cell according to any one of claims 1 to 21, 49 or 50, the polypeptide according to any one of claims 34 to 45, a polynucleotide according to claim 46, a vector of claim 47 or claim 48, an oil according to any one of claims 56 to 65, a fatty acid of claim 66 and/or a feedstuff of claim 69.
73. The method of claim 72, wherein the condition is cardiac arrhythmia's, angioplasty, inflammation, asthma, psoriasis, osteoporosis, kidney stones, AIDS, multiple sclerosis, rheumatoid arthritis, Crohn's disease, schizophrenia, cancer, foetal alcohol syndrome, attention deficient hyperactivity disorder, cystic fibrosis, phenylketonuria, unipolar depression, aggressive hostility, adrenoleukodystophy, coronary heart disease, hypertension, diabetes, obesity, Alzheimer's disease, chronic obstructive pulmonary disease, ulcerative colitis, restenosis after angioplasty, eczema, high blood pressure, platelet aggregation, gastrointestinal bleeding, endometriosis, premenstrual syndrome, myalgic encephalomyelitis, chronic fatigue after viral infections or an ocular disease.
74. Use of a cell according to any one of claims 1 to 21, 49 or 50, the polypeptide according to any one of claims 34 to 45, a polynucleotide according to claim 46, a vector of claim 47 or claim 48, an oil according to any one of claims 56 to 65 or a fatty acid of claim 66 and/or a feedstuff of claim 69 for the manufacture of a medicament for treating or preventing a condition which would benefit from a PUFA.
PCT/AU2007/001242 2006-08-29 2007-08-29 Synthesis of fatty acids WO2008025068A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
AU2007291937A AU2007291937B2 (en) 2006-08-29 2007-08-29 Synthesis of fatty acids
CA002661697A CA2661697A1 (en) 2006-08-29 2007-08-29 Synthesis of fatty acids
BRPI0716075-5A BRPI0716075A2 (en) 2006-08-29 2007-08-29 Fatty Acid Synthesis
US12/310,645 US8816106B2 (en) 2006-08-29 2007-08-29 Synthesis of fatty acids
EP07784864A EP2059588A4 (en) 2006-08-29 2007-08-29 Synthesis of fatty acids
US14/467,942 US10513717B2 (en) 2006-08-29 2014-08-25 Synthesis of fatty acids

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US84128506P 2006-08-29 2006-08-29
US60/841,285 2006-08-29

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/310,645 A-371-Of-International US8816106B2 (en) 2006-08-29 2007-08-29 Synthesis of fatty acids
US14/467,942 Continuation US10513717B2 (en) 2006-08-29 2014-08-25 Synthesis of fatty acids

Publications (1)

Publication Number Publication Date
WO2008025068A1 true WO2008025068A1 (en) 2008-03-06

Family

ID=39135404

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2007/001242 WO2008025068A1 (en) 2006-08-29 2007-08-29 Synthesis of fatty acids

Country Status (8)

Country Link
US (2) US8816106B2 (en)
EP (1) EP2059588A4 (en)
CN (1) CN101578363A (en)
AU (1) AU2007291937B2 (en)
BR (1) BRPI0716075A2 (en)
CA (1) CA2661697A1 (en)
RU (1) RU2009111266A (en)
WO (1) WO2008025068A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012003545A1 (en) * 2010-07-09 2012-01-12 Commonwealth Scientific And Industrial Research Organisation Acetylenation of fatty acids
US8530724B2 (en) 2006-07-14 2013-09-10 Commonwealth Scientific And Industrial Research Organisation Altering the fatty acid composition of rice
US8716555B2 (en) 2008-07-21 2014-05-06 Commonwealth Scientific And Industrial Research Organisation Cottonseed oil and uses
US8735111B2 (en) 2011-12-27 2014-05-27 Commonwealth Scientific And Industrial Research Organisation Processes for producing hydrocarbon products
US8765404B2 (en) 2010-03-02 2014-07-01 Massachusetts Institute Of Technology Microbial engineering for the production of fatty acids and fatty acid derivatives
US8809026B2 (en) 2011-12-27 2014-08-19 Commonwealth Scientific And Industrial Research Organisation Processes for producing lipids
US8853432B2 (en) 2004-04-22 2014-10-07 Commonwealth Scientific And Industrial Research Organisation Synthesis of long-chain polyunsaturated fatty acids by recombinant cell
US8921652B2 (en) 2008-07-21 2014-12-30 Commonwealth Scientific And Industrial Research Organisation Vegetable oils and uses therefor
US8946460B2 (en) 2012-06-15 2015-02-03 Commonwealth Scientific And Industrial Research Organisation Process for producing polyunsaturated fatty acids in an esterified form
US9127288B2 (en) 2010-06-28 2015-09-08 Commonwealth Scientific And Industrial Research Organisation Methods of producing lipids
US9453183B2 (en) 2004-04-22 2016-09-27 Commonwealth Scientific And Industrial Research Organisation Synthesis of long-chain polyunsaturated fatty acids by recombinant cell
US9718759B2 (en) 2013-12-18 2017-08-01 Commonwealth Scientific And Industrial Research Organisation Lipid comprising docosapentaenoic acid
US9938486B2 (en) 2008-11-18 2018-04-10 Commonwealth Scientific And Industrial Research Organisation Enzymes and methods for producing omega-3 fatty acids
US10005713B2 (en) 2014-06-27 2018-06-26 Commonwealth Scientific And Industrial Research Organisation Lipid compositions comprising triacylglycerol with long-chain polyunsaturated fatty acids at the sn-2 position
US10323209B2 (en) 2012-04-25 2019-06-18 Commonwealth Scientific And Industrial Research Organisation High oleic acid oils
US10472587B2 (en) 2014-07-07 2019-11-12 Commonwealth Scientific And Industrial Research Organisation Processes for producing industrial products from plant lipids
US10513717B2 (en) 2006-08-29 2019-12-24 Commonwealth Scientific And Industrial Research Organisation Synthesis of fatty acids
US11639507B2 (en) 2011-12-27 2023-05-02 Commonwealth Scientific And Industrial Research Organisation Processes for producing lipids
US11859193B2 (en) 2016-09-02 2024-01-02 Nuseed Global Innovation Ltd. Plants with modified traits

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009129583A1 (en) * 2008-04-25 2009-10-29 Commonwealth Scientific Industrial Research Organisation Recombinant cells and methods for hydroxylating fatty acids
CA2722275A1 (en) * 2008-04-25 2009-10-29 Commonwealth Scientific And Industrial Research Organisation Polypeptides and methods for producing triacylglycerols comprising modified fatty acids
US11718577B2 (en) 2013-12-18 2023-08-08 Commonwealth Scientific And Industrial Research Organisation Lipid compositions comprising triacylglycerol with long-chain polyunsaturated fatty acids
EP3097910B1 (en) * 2014-01-24 2020-05-13 Kyoto University Anti-inflammatory agent containing rare fatty acid
CN107494989A (en) * 2017-07-21 2017-12-22 浙江海洋大学 The man-made feeds formula and processing method of a kind of hickie cuttlefish
CN107494362A (en) * 2017-07-21 2017-12-22 浙江海洋大学 A kind of bait intensifying method for improving hickie squid larva survival rate
CN107535743A (en) * 2017-07-21 2018-01-05 浙江海洋大学 One kind is suitable to true octopus floating larva culture fortification formula
CN107484921A (en) * 2017-08-24 2017-12-19 浦江县协盈动物饲料技术开发有限公司 The preparation method of shallow sea fish fry phase mixed feed
CN107616355A (en) * 2017-08-24 2018-01-23 浦江县协盈动物饲料技术开发有限公司 The preparation method of grouper nutrition fortifier
CN107616354A (en) * 2017-08-24 2018-01-23 浦江县协盈动物饲料技术开发有限公司 Increase the preparation method of the shallow sea fish feed additive of Meat Tenderness
CN107821743A (en) * 2017-10-13 2018-03-23 宁波市鄞州区超锐食品科技有限公司 Catfish scrod feed
CN107874015A (en) * 2017-10-13 2018-04-06 宁波市鄞州区超锐食品科技有限公司 Freshwater aquaculture fish feed attractant growth-promoting additive
CN108185214A (en) * 2017-11-30 2018-06-22 兰溪市酉泽饲料技术服务有限公司 Steamed crab is promoted to change the preparation method of the feed of shell
US11913006B2 (en) 2018-03-16 2024-02-27 Nuseed Global Innovation Ltd. Plants producing modified levels of medium chain fatty acids
CN114028542A (en) * 2021-12-13 2022-02-11 中科华南生命科学(深圳)研究中心 Eye disease treatment formula with multiple repair factors
CN114426990B (en) * 2022-03-07 2023-08-11 湖北师范大学 Gao Yadi acid salt tolerant bacteria mediated synthesis biological tellurium nano-particle and antibacterial application thereof

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5004863A (en) 1986-12-03 1991-04-02 Agracetus Genetic engineering of cotton plants and lines
WO1991013980A1 (en) 1990-03-16 1991-09-19 Calgene, Inc. Novel sequences preferentially expressed in early seed development and methods related thereto
AU6178194A (en) 1993-03-11 1994-09-26 National Research Council Of Canada Enhanced regeneration system for cereals
CA2092588A1 (en) 1993-03-26 1994-09-27 Narender S. Nehra Enhanced regeneration system for cereals
US5416011A (en) 1988-07-22 1995-05-16 Monsanto Company Method for soybean transformation and regeneration
US5463174A (en) 1986-05-29 1995-10-31 Calgene Inc. Transformation and foreign gene expression in Brassica species
US5504200A (en) 1983-04-15 1996-04-02 Mycogen Plant Science, Inc. Plant gene expression
AU667939B2 (en) 1992-07-07 1996-04-18 Japan Tobacco Inc. Method of transforming monocotyledon
US5518908A (en) 1991-09-23 1996-05-21 Monsanto Company Method of controlling insects
WO1997006269A1 (en) 1995-08-03 1997-02-20 Zeneca Limited Inducible herbicide resistance
US5608152A (en) 1986-07-31 1997-03-04 Calgene, Inc. Seed-specific transcriptional regulation
WO1998045461A1 (en) 1997-04-09 1998-10-15 Rhone-Poulenc Agro An oleosin 5' regulatory region for the modification of plant seed lipid composition
WO1999014314A1 (en) 1997-09-12 1999-03-25 Commonwealth Scientific And Industrial Research Organisation Regulation of gene expression in plants
WO1999049050A2 (en) * 1998-03-20 1999-09-30 E.I. Du Pont De Nemours And Company Limanthes oil genes
US6100447A (en) 1998-02-12 2000-08-08 Applied Phytologics, Inc. Method of barley transformation
WO2001096363A1 (en) * 2000-06-12 2001-12-20 Boyce Thompson Institute For Plant Research Fatty acid desaturase gene and protein for modulating activation of defense signaling pathways in plants
US6432684B1 (en) 1997-04-11 2002-08-13 Abbott Laboratories Human desaturase gene and uses thereof
US20030033633A1 (en) 1992-11-17 2003-02-13 Lightner Jonathan Edward Genes for microsomal delta-12 fatty acid desaturases and related enzymes from plants
US6541257B2 (en) 1997-04-29 2003-04-01 The Regents Of The University Of California Method for preparing barley green regenerative tissue
US7109392B1 (en) * 1996-10-09 2006-09-19 Cargill, Incorporated Methods for increasing oleic acid content in seeds from transgenic plants containing a mutant delta 12 desaturase
US20070238648A1 (en) * 2000-09-26 2007-10-11 Brownlie Alison J Methods and compositions employing a novel stearoyl-CoA desaturase-hSCD5
US9710621B1 (en) 2011-11-02 2017-07-18 Google Inc. Platform for cloud application software

Family Cites Families (187)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2002178A (en) 1933-01-16 1935-05-21 Floyd S Warring Sprinkler
US7135614B1 (en) 1990-08-30 2006-11-14 Cargill, Incorporated Brassica or helianthus plants having mutant delta-12 or delta-15 sequences
US5861187A (en) 1990-08-30 1999-01-19 Cargill, Incorporated Oil from canola seed with altered fatty acid profiles and a method of producing oil
US6683232B1 (en) 1991-10-10 2004-01-27 Rhone-Poulenc Agrochimie Production of γ linolenic acid by a Δ6-desaturase
PH31293A (en) 1991-10-10 1998-07-06 Rhone Poulenc Agrochimie Production of y-linolenic acid by a delta6-desaturage.
US6355861B1 (en) 1991-10-10 2002-03-12 Rhone-Poulenc Agrochimie Production of gamma linolenic acid by a Δ6-desaturase
US5614393A (en) 1991-10-10 1997-03-25 Rhone-Poulenc Agrochimie Production of γ-linolenic acid by a Δ6-desaturase
DK0616644T3 (en) 1991-12-04 2003-10-27 Du Pont Fatty acid desaturase genes from plants
AU673359B2 (en) 1992-05-15 1996-11-07 Sagami Chemical Research Center Gene which codes for eicosapentaenoic acid synthetase group and process for producing eicosapentaenoic acid
US5683898A (en) 1992-05-15 1997-11-04 Sagami Chemical Research Center Gene coding for eicosapentaenoic acid synthesizing enzymes and process for production of eicosapentaenoic acid
US5798259A (en) 1992-05-15 1998-08-25 Sagami Chemical Research Center Gene coding for eicosapentaenoic acid synthesizing enzymes and process for production of eicosapentaenoic acid
US6372965B1 (en) * 1992-11-17 2002-04-16 E.I. Du Pont De Nemours And Company Genes for microsomal delta-12 fatty acid desaturases and hydroxylases from plants
AU750363B2 (en) 1997-06-12 2002-07-18 Cargill Incorporated Fatty acid desaturases and mutant sequences thereof
US7541519B2 (en) 1995-12-14 2009-06-02 Cargill, Incorporated Fatty acid desaturases and mutant sequences thereof
ATE319296T1 (en) * 1995-12-14 2006-03-15 Cargill Inc PLANTS WITH MUTATED SEQUENCES WHICH PROVIDE ALTERED FATTY ACID CONTENT
US6342658B1 (en) 1995-12-14 2002-01-29 Cargill, Incorporated Fatty acid desaturases and mutant sequences thereof
EP0913473A4 (en) 1996-07-10 2000-12-06 Sagami Chem Res Process for producing icosapentaenoic acid by genetic recombination
WO1998018952A1 (en) 1996-10-30 1998-05-07 Nippon Suisan Kaisha, Ltd. Process for producing fats containing highly unsaturated fatty acids containing selectively concentrated docosahexaenoic acid
US6194167B1 (en) 1997-02-18 2001-02-27 Washington State University Research Foundation ω-3 fatty acid desaturase
JP4127860B2 (en) 1997-03-04 2008-07-30 サントリー株式会社 Polyunsaturated fatty acid and method for producing lipid containing the same
US6428990B1 (en) 1997-04-11 2002-08-06 Abbott Laboratories Human desaturase gene and uses thereof
US6051754A (en) 1997-04-11 2000-04-18 Abbott Laboratories Methods and compositions for synthesis of long chain poly-unsaturated fatty acids in plants
US5968809A (en) 1997-04-11 1999-10-19 Abbot Laboratories Methods and compositions for synthesis of long chain poly-unsaturated fatty acids
ATE297994T1 (en) 1997-04-11 2005-07-15 Calgene Llc METHODS AND COMPOSITIONS FOR THE SYNTHESIS OF LONG-CHAIN POLYUNSATURATED FATTY ACIDS IN PLANTS.
US6075183A (en) 1997-04-11 2000-06-13 Abbott Laboratories Methods and compositions for synthesis of long chain poly-unsaturated fatty acids in plants
US5972664A (en) 1997-04-11 1999-10-26 Abbott Laboratories Methods and compositions for synthesis of long chain poly-unsaturated fatty acids
US7589253B2 (en) 1997-04-15 2009-09-15 Commonwealth Scientific And Industrial Research Organisation Fatty acid epoxygenase genes from plants and uses therefor in modifying fatty acid metabolism
IL133272A0 (en) 1997-06-04 2001-04-30 Calgene Llc Production of polyunsaturated fatty acid by expression of polyketide-like synthesis genes in plants
US6566583B1 (en) 1997-06-04 2003-05-20 Daniel Facciotti Schizochytrium PKS genes
GB9724783D0 (en) 1997-11-24 1998-01-21 Inst Arable Crops Research Novel polypeptides
KR20010033517A (en) 1997-12-23 2001-04-25 더 유니버시티 오브 브리스톨 Desaturase
US20030152983A1 (en) 1997-12-23 2003-08-14 University Of Bristol Desaturase
US6838594B1 (en) 1998-03-20 2005-01-04 E. I. Du Pont De Nemours And Company Limnanthes oil genes
US6492108B1 (en) 1998-03-26 2002-12-10 Incyte Genomics, Inc. Delta-6 desaturase homologs
JP2002516093A (en) 1998-05-29 2002-06-04 オハイオ ユニバーシティー Compositions and methods for synthesizing fatty acids, their derivatives and downstream products
WO1999064616A2 (en) 1998-06-12 1999-12-16 Abbott Laboratories Polyunsaturated fatty acids in plants
DK1086236T3 (en) 1998-06-12 2007-12-03 Calgene Llc Polyunsaturated fatty acids in plants
HUP0103694A3 (en) 1998-08-20 2003-12-29 Du Pont Genes for plant fatty acid modifiyng enzymes associated with conjugated double bond formation
WO2000011012A1 (en) 1998-08-24 2000-03-02 Rutgers, The State University Of New Jersey Synthetic fatty acid desaturase gene for expression in plants
US6913916B1 (en) 1998-09-02 2005-07-05 Abbott Laboratories Elongase genes and uses thereof
US6403349B1 (en) 1998-09-02 2002-06-11 Abbott Laboratories Elongase gene and uses thereof
US6677145B2 (en) 1998-09-02 2004-01-13 Abbott Laboratories Elongase genes and uses thereof
US20030163845A1 (en) 1998-09-02 2003-08-28 Pradip Mukerji Elongase genes and uses thereof
WO2000020603A1 (en) 1998-10-05 2000-04-13 Abbott Laboratories Altered fatty acid biosynthesis in insect cells using delta five desaturase
AU1097900A (en) 1998-10-05 2000-04-26 Abbott Laboratories Delta 6 and delta 12 desaturases and modified fatty acid biosynthesis and products produced therefrom
JP2002527051A (en) 1998-10-09 2002-08-27 メルク エンド カムパニー インコーポレーテッド Delta-6 fatty acid desaturase
US6197294B1 (en) 1998-10-26 2001-03-06 Neurotech S.A. Cell surface molecule-induced macrophage activation
JP4182192B2 (en) 1998-11-26 2008-11-19 独立行政法人産業技術総合研究所 Genes encoding enzymes involved in biosynthesis of icosapentaenoic acid and / or docosahexaenoic acid
US6864077B1 (en) 1998-12-03 2005-03-08 Edgar B. Cahoon Membrane-bound desaturases
JP2001095588A (en) 1998-12-04 2001-04-10 Research Institute Of Innovative Technology For The Earth Gene of desaturase for omega-3 fatty acid of green alga and its use
US6825017B1 (en) 1998-12-07 2004-11-30 Washington State University Research Foundation Desaturases and methods of using them for synthesis of polyunsaturated fatty acids
US8003772B2 (en) 1999-01-14 2011-08-23 Martek Biosciences Corporation Chimeric PUFA polyketide synthase systems and uses thereof
US7217856B2 (en) 1999-01-14 2007-05-15 Martek Biosciences Corporation PUFA polyketide synthase systems and uses thereof
US7247461B2 (en) 1999-01-14 2007-07-24 Martek Biosciences Corporation Nucleic acid molecule encoding ORFA of a PUFA polyketide synthase system and uses thereof
US7271315B2 (en) 1999-01-14 2007-09-18 Martek Biosciences Corporation PUFA polyketide synthase systems and uses thereof
US20070244192A1 (en) 1999-01-14 2007-10-18 Martek Biosciences Corporation Plant seed oils containing polyunsaturated fatty acids
US7211418B2 (en) 1999-01-14 2007-05-01 Martek Biosciences Corporation PUFA polyketide synthase systems and uses thereof
AU3516100A (en) 1999-03-05 2000-09-21 Monsanto Technology Llc Multigene expression vectors for the biosynthesis of products via multienzyme biological pathways
EP1035207A1 (en) 1999-03-09 2000-09-13 MultiGene Biotech GmbH cDNA molecules of the members of gene family encoding human fatty acid desaturases and their use in diagnosis and therapy
CA2365096A1 (en) 1999-03-18 2000-09-21 The University Of Bristol Polyunsaturated fatty acid (pufa) elongase from caenorhabditis elegans
DK1181373T3 (en) 1999-06-07 2006-11-27 Basf Plant Science Gmbh Delta-6-acetylenase and delta-6-desaturase from Ceratodon purpureus
CA2378423A1 (en) 1999-07-06 2001-01-11 Basf Plant Science Gmbh Plants expressing .delta.6-desaturase genes and oils from these plants containing pufas and method for producing unsaturated fatty acids
WO2001004636A1 (en) 1999-07-12 2001-01-18 Ohio University Mammalian cells expressing desaturases and elongases
US7070970B2 (en) 1999-08-23 2006-07-04 Abbott Laboratories Elongase genes and uses thereof
US7531718B2 (en) 1999-08-26 2009-05-12 Monsanto Technology, L.L.C. Nucleic acid sequences and methods of use for the production of plants with modified polyunsaturated fatty acids
ATE383433T1 (en) 1999-08-26 2008-01-15 Calgene Llc PLANTS WITH MODIFIED POLYUNSATURATED FATTY ACIDS
US7067722B2 (en) 1999-08-26 2006-06-27 Monsanto Technology Llc Nucleic acid sequences and methods of use for the production of plants with modified polyunsaturated fatty acids
AU7285300A (en) 1999-09-10 2001-04-17 Celanese Ventures Gmbh Nucleic acid which is obtained from tetrahymena and which codes for a delta-6-desaturase, the production thereof and use
FR2798391B1 (en) 1999-09-15 2001-12-14 Inst Nat Sante Rech Med THYROID ENZYME NADPH OXYDASE, NUCLEIC ACID ENCODING THIS ENZYME AND THEIR APPLICATIONS
JP4221476B2 (en) 1999-11-19 2009-02-12 独立行政法人産業技術総合研究所 Plasmid cloned icosapentaenoic acid biosynthesis genes and cyanobacteria producing icosapentaenoic acid
IL149388A0 (en) 1999-11-22 2002-11-10 Plant Bioscience Ltd Enhanced transgene expression by co-expression with a suppressor of post-transcriptional gene silencing (ptgs)
WO2001038541A1 (en) 1999-11-25 2001-05-31 Basf Plant Science Gmbh Moss genes from physcomitrella patents encoding proteins involved in the synthesis of polyunsaturated fatty acids and lipids
GB9929681D0 (en) 1999-12-15 2000-02-09 Implyx Ltd Transgenic insect
JP2001169780A (en) 1999-12-15 2001-06-26 Natl Inst Of Advanced Industrial Science & Technology Meti Gene derived from docosahexaenoic acid-producing bacterium
GB9929897D0 (en) 1999-12-18 2000-02-09 Slabas Antoni R Improvements in or relating to conjugated fatty acids and related compounds
US20030172398A1 (en) 1999-12-21 2003-09-11 Browse John A. Novel delta-12 desaturase and methods of using it for synthesis of polyunsaturated fatty acids
ES2331228T3 (en) 2000-02-09 2009-12-28 Basf Se NEW ELONGASA GENE AND PROCESS FOR THE PREPARATION OF POLYINSATURATED FATTY ACIDS.
US6686185B1 (en) 2000-03-07 2004-02-03 Millennium Pharmaceuticals, Inc. 25934, a novel fatty acid desaturase and uses therefor
US20040157221A9 (en) 2000-03-07 2004-08-12 Millennium Pharmaceuticals, Inc. Novel 25869, 25934, 26335, 50365, 21117, 38692, 46508, 16816, 16839, 49937, 49931 and 49933 molecules and uses therefor
US20020111307A1 (en) 2000-06-23 2002-08-15 Glucksmann Maria Alexandra 46508, a novel human peptidyl-tRNA hydrolase family member and uses thereof
US7411054B2 (en) 2000-03-07 2008-08-12 Millennium Pharmaceuticals, Inc. 25869, 25934, 26335, 50365, 21117, 38692, 46508, 16816, 16839, 49937, 49931 and 49933 molecules and uses therefor
WO2001073060A2 (en) 2000-03-24 2001-10-04 Millennium Pharmaceuticals, Inc. 18221, dual specificity phosphatase and uses thereof
CA2301158A1 (en) 2000-03-24 2001-09-24 Stephen J. Allen Screening methods for compounds useful for modulating lipid metabolism in disease
WO2001075069A1 (en) 2000-03-31 2001-10-11 Idemitsu Petrochemical Co., Ltd. Process for producing lipids and lipid-secreting microorganisms
US7091005B2 (en) 2000-05-16 2006-08-15 Merck & Co., Inc. Gene responsible for Stargardt-like dominant macular dystrophy
US20040195512A1 (en) 2000-05-16 2004-10-07 Crosetto Dario B. Method and apparatus for anatomical and functional medical imaging
EP1404820A2 (en) 2000-05-19 2004-04-07 Millennium Pharmaceuticals, Inc. 50365, a hexokinase family member and uses therefor
AU2001263473A1 (en) 2000-05-26 2001-12-11 Washington State University Research Foundation Palmitate desaturase gene
WO2002002760A2 (en) 2000-06-29 2002-01-10 Millennium Pharmaceuticals, Inc. 25869, a human carboxylesterase and uses thereof
AU2001275990A1 (en) 2000-07-17 2002-01-30 Millennium Pharmaceuticals, Inc. 16816 and 16839, novel human phospholipase c molecules and uses therefor
GB0316629D0 (en) 2003-07-16 2003-08-20 Univ York Transgenic cell
AU2001286621A1 (en) 2000-08-21 2002-03-04 Millennium Pharmaceuticals, Inc. Human transporter family members and uses thereof
PT1911837E (en) 2000-09-28 2011-06-07 Bioriginal Food & Science Corp Fad5-2 fatty acid desaturase family member and uses thereof
AU2140402A (en) 2000-11-29 2002-06-11 Xenon Genetics Inc Human elongase genes uses thereof and compounds for modulating same
DE10102338A1 (en) 2001-01-19 2002-07-25 Basf Plant Science Gmbh New expression cassette for plant genes, useful for preparing transgenic plants that have increased production of polyunsaturated fatty acids
DE10102337A1 (en) 2001-01-19 2002-07-25 Basf Plant Science Gmbh Preparing ester containing polyunsaturated fatty acids, useful e.g. in animal or human nutrition, by transforming organism with desaturase gene from Phaeodactylum tricornatum
US6635451B2 (en) 2001-01-25 2003-10-21 Abbott Laboratories Desaturase genes and uses thereof
JP4347572B2 (en) 2001-01-25 2009-10-21 アボット・ラボラトリーズ Desaturase gene and its use
DE10106660A1 (en) 2001-02-12 2002-08-29 Celanese Ventures Gmbh Process for the production of gamma-linolenic acid from a ciliate culture by adding suitable precursor molecules to the culture medium
GB0107510D0 (en) 2001-03-26 2001-05-16 Univ Bristol New elongase gene and a process for the production of -9-polyunsaturated fatty acids
WO2002081702A1 (en) 2001-04-05 2002-10-17 Idemitsu Technofine Co., Ltd. Fatty acid δ6-position desaturase genes and plasmids and tra nsformant containing these genes
TWI350854B (en) 2001-04-16 2011-10-21 Martek Biosciences Corp Product and process for transformation of thraustochytriales microorganisms
TWI337619B (en) 2001-04-16 2011-02-21 Martek Biosciences Corp Pufa polyketide synthase systems and uses thereof
US20030082754A1 (en) 2001-05-04 2003-05-01 Pradip Mukerji Delta4 - desaturase genes and uses thereof
US7045683B2 (en) 2001-05-04 2006-05-16 Abbott Laboratories Δ4-desaturase genes and uses thereof
EP2255668A3 (en) 2001-05-14 2012-04-04 Martek Biosciences Corporation Production and Use of a Polar Lipid-Rich Fraction Containing Omega-3 and/or Omega-6 Highly Unsaturated Fatty Acids from Microbes, Genetically Modified Plant Seeds and Marine Organisms
DE10134660A1 (en) 2001-07-20 2003-02-06 Basf Plant Science Gmbh New isolated nucleic acid encoding desaturase enzymes from pomegranate, useful for recombinant production of unsaturated fatty acids, for e.g. the production of food, animal feeds and pharmaceuticals
US20050089879A1 (en) 2001-07-31 2005-04-28 Ivo Feussner Method for producing arachidonic acid in transgenic organisms
US6875595B2 (en) 2001-09-13 2005-04-05 Divergence, Inc. Nematode fatty acid desaturase-like sequences
JP2003116566A (en) 2001-10-12 2003-04-22 Osamu Suzuki METHOD FOR PRODUCING n-3-BASED DOCOSAPENTAENOIC ACID
DE50312241D1 (en) 2002-01-30 2010-01-28 Basf Plant Science Gmbh METHOD FOR PRODUCING MULTIPLE UNSATURATED F
US7211656B2 (en) 2002-01-30 2007-05-01 Abbott Laboratories Desaturase genes, enzymes encoded thereby, and uses thereof
GB2385852A (en) 2002-02-27 2003-09-03 Rothamsted Ex Station Delta 6-desaturases from Primulaceae
US7700833B2 (en) 2002-03-01 2010-04-20 Cornell University Process for the production of unsaturated fatty acids
US7705202B2 (en) 2002-03-16 2010-04-27 The University Of York Transgenic plants expressing enzymes involved in fatty acid biosynthesis
DE10219203A1 (en) 2002-04-29 2003-11-13 Basf Plant Science Gmbh Process for the production of polyunsaturated fatty acids in plants
BRPI0311324B1 (en) 2002-05-22 2020-01-28 Monsanto Technology Llc polynucleotide, vector, polypeptide with desasturase activity, methods for plant production and corn oil containing omega-3 fatty acids, to increase the nutritional value of an edible product, food or feed manufacture, as well as edible composition
AU2003258496A1 (en) 2002-07-03 2004-01-23 Basf Plant Science Gmbh Method for the production of conjugated polyunsaturated fatty acids comprising at least two double bonds in plants
WO2004057001A2 (en) 2002-12-19 2004-07-08 University Of Bristol Method for the production of polyunsaturated fatty acids
US8084074B2 (en) 2003-02-12 2011-12-27 E. I. Du Pont De Nemours And Company Production of very long chain polyunsaturated fatty acids in oil seed plants
US20040172682A1 (en) 2003-02-12 2004-09-02 Kinney Anthony J. Production of very long chain polyunsaturated fatty acids in oilseed plants
WO2004076617A2 (en) 2003-02-27 2004-09-10 Basf Plant Science Gmbh Method for the production of polyunsaturated fatty acids
CA2868312A1 (en) 2003-03-31 2004-10-14 University Of Bristol Novel plant acyltransferases specific for long-chained, multiply unsaturated fatty acids
DE10315026A1 (en) 2003-04-02 2004-10-14 Bioplanta Arzneimittel Gmbh Compositions containing omega-3 fatty acid oils and plant extracts
US7629503B2 (en) 2003-04-08 2009-12-08 Basf Plant Science Gmbh Δ-4 desaturases from Euglena gracilis, expressing plants, and oils containing PUFA
EP1618193B1 (en) 2003-04-16 2008-05-07 BASF Plant Science GmbH Use of genes for increasing the oil content in plants
US7125672B2 (en) 2003-05-07 2006-10-24 E. I. Du Pont De Nemours And Company Codon-optimized genes for the production of polyunsaturated fatty acids in oleaginous yeasts
US8313911B2 (en) 2003-05-07 2012-11-20 E I Du Pont De Nemours And Company Production of polyunsaturated fatty acids in oleaginous yeasts
US20110059496A1 (en) 2003-06-25 2011-03-10 E. I. Du Pont De Nemours And Company Glyceraldehyde-3-phosphate dehydrogenase and phosphoglycerate mutase promoters for gene expression in oleaginous yeast
US7267976B2 (en) 2003-07-02 2007-09-11 E.I. Du Pont De Nemours And Company Acyltransferases for alteration of polyunsaturated fatty acids and oil content in oleaginous yeasts
BRPI0413073A (en) 2003-08-01 2006-10-03 Basf Plant Science Gmbh process for the production of compounds, oil, lipid or fatty acid, or a fraction thereof, oil, lipid or fatty acid composition, process for the production of oils, lipids or fatty acid compositions, use of oil, lipid or acids fatty acids or compositions of oil, lipid or fatty acid or oils, lipids or fatty acid compositions, isolated nucleic acid sequence, amino acid sequence, gene construct, vector, and transgenic non-human organism
ATE430802T1 (en) 2003-08-21 2009-05-15 Monsanto Technology Llc FATTY ACID DESATURASES FROM PRIMULA
AU2004290052B2 (en) 2003-11-12 2008-12-04 Corteva Agriscience Llc Delta-15 desaturases suitable for altering levels of polyunsaturated fatty acids in oleaginous plants and yeast
US7504259B2 (en) 2003-11-12 2009-03-17 E. I. Du Pont De Nemours And Company Δ12 desaturases suitable for altering levels of polyunsaturated fatty acids in oleaginous yeast
MY140210A (en) 2003-12-22 2009-11-30 Suntory Holdings Ltd Marchantiales-derived unsaturated fatty acid synthetase genes and use of the same
CA2556336C (en) 2004-02-17 2015-09-08 The University Of York Desaturase enzymes
JP4567047B2 (en) 2004-02-27 2010-10-20 ビーエーエスエフ プラント サイエンス ゲーエムベーハー Process for producing polyunsaturated fatty acids in transgenic plants
ATE528403T1 (en) 2004-02-27 2011-10-15 Basf Plant Science Gmbh METHOD FOR PRODUCING UNSATURATED OMEGA-3 FATTY ACIDS IN TRANSGENIC ORGANISMS
WO2005083093A2 (en) 2004-02-27 2005-09-09 Basf Plant Science Gmbh Method for producing polyunsaturated fatty acids in transgenic plants
DE102004017369A1 (en) 2004-04-08 2005-11-03 Nutrinova Nutrition Specialties & Food Ingredients Gmbh Screening method for the identification of PUFA-PKS in samples
DE102004017370A1 (en) 2004-04-08 2005-10-27 Nutrinova Nutrition Specialties & Food Ingredients Gmbh PUFA-PKS Genes from Ulkenia
US8378186B2 (en) 2004-04-16 2013-02-19 Monsanto Technology Llc Expression of fatty acid desaturases in corn
CA2563875C (en) 2004-04-22 2015-06-30 Commonwealth Scientific And Industrial Research Organisation Synthesis of long-chain polyunsaturated fatty acids by recombinant cells
US7807849B2 (en) 2004-04-22 2010-10-05 Commonwealth Scientific And Industrial Research Organisation Synthesis of long-chain polyunsaturated fatty acids by recombinant cells
BRPI0511236A (en) 2004-06-04 2007-11-27 Fluxome Sciences As A method for producing four or more double bond polyunsaturated fatty acids, genetically modified saccharomyces cerevisiae which is capable of producing four or more double bond polyunsaturated fatty acids when developed on a substrate other than fatty acid, composition, use of the composition and use of a genetically modified saccharomyces cervisiae
BRPI0512481A (en) 2004-06-25 2008-03-11 Du Pont isolated polynucleotide, polypeptide, recombinant construct, cell, transformed yarrowia sp.transformed, methods for transforming a cell, producing a transformed plant, producing yeast, producing polyunsaturated fatty acids and producing at least one polyunsaturated fatty acid, seeds, oils, oilseed plants and food or feed
DE102004060340A1 (en) 2004-07-16 2006-02-09 Basf Plant Science Gmbh Method for increasing the content of polyunsaturated long-chain fatty acids in transgenic organisms
US7270315B2 (en) 2004-09-28 2007-09-18 Amt International, Inc. Exchange column, perforated tray assembly
GB0421937D0 (en) 2004-10-02 2004-11-03 Univ York Acyl CoA synthetases
US8685679B2 (en) 2004-11-04 2014-04-01 E I Du Pont De Nemours And Company Acyltransferase regulation to increase the percent of polyunsaturated fatty acids in total lipids and oils of oleaginous organisms
US7192762B2 (en) 2004-11-04 2007-03-20 E. I. Du Pont De Nemours And Company Mortierella alpina glycerol-3-phosphate o-acyltransferase for alteration of polyunsaturated fatty acids and oil content in oleaginous organisms
US20060094102A1 (en) 2004-11-04 2006-05-04 Zhixiong Xue Ammonium transporter promoter for gene expression in oleaginous yeast
US7588931B2 (en) 2004-11-04 2009-09-15 E. I. Du Pont De Nemours And Company High arachidonic acid producing strains of Yarrowia lipolytica
US7198937B2 (en) 2004-11-04 2007-04-03 E. I. Du Pont De Nemours And Company Mortierella alpina diacylglycerol acyltransferase for alteration of polyunsaturated fatty acids and oil content in oleaginous organisms
US7273746B2 (en) 2004-11-04 2007-09-25 E.I. Dupont De Nemours And Company Diacylglycerol acyltransferases for alteration of polyunsaturated fatty acids and oil content in oleaginous organisms
US7189559B2 (en) 2004-11-04 2007-03-13 E. I. Du Pont De Nemours And Company Mortierella alpina lysophosphatidic acid acyltransferase homolog for alteration of polyunsaturated fatty acids and oil content in oleaginous organisms
CN101124330A (en) 2004-12-14 2008-02-13 阿维斯塔金格兰技术有限公司 Recombinant production docosahexaenoic acid (DHA) in yeast
DE102004063326A1 (en) 2004-12-23 2006-07-06 Basf Plant Science Gmbh Process for producing polyunsaturated fatty acids in transgenic organisms
DE102004062294A1 (en) * 2004-12-23 2006-07-06 Basf Plant Science Gmbh Process for the preparation of polyunsaturated long-chain fatty acids in transgenic organisms
DE102005013779A1 (en) 2005-03-22 2006-09-28 Basf Plant Science Gmbh Process for the preparation of polyunsaturated C20 and C22 fatty acids having at least four double bonds in transgenic plants
WO2006135866A2 (en) 2005-06-10 2006-12-21 Martek Biosciences Corporation Pufa polyketide synthase systems and uses thereof
WO2007005882A2 (en) 2005-07-05 2007-01-11 North Carolina State University Methods and compositions for expressing proteins in plants
US7645604B2 (en) 2005-11-23 2010-01-12 E.I. Du Pont De Nemours And Company Delta-9 elongases and their use in making polyunsaturated fatty acids
EP1981331A4 (en) 2006-02-03 2009-09-09 Texas A & M Univ Sys Enhancing expression of value-added genes by transgenic expression of tombusvirus-based p19 gene mutants
AR059376A1 (en) 2006-02-21 2008-03-26 Basf Plant Science Gmbh PROCEDURE FOR THE PRODUCTION OF POLYINSATURATED FATTY ACIDS
ES2527875T3 (en) 2006-03-15 2015-02-02 Dsm Ip Assets B.V. Production of polyunsaturated fatty acids in heterologous organisms using AGPI polyketide synthase systems
US7943823B2 (en) 2006-04-28 2011-05-17 E.I. Du Pont De Nemours And Company Delta-8 desaturase and its use in making polyunsaturated fatty acids
JP4230494B2 (en) 2006-06-06 2009-02-25 日立建機株式会社 Electric drive truck drive system
DE102006034313A1 (en) 2006-07-21 2008-01-24 Basf Plant Science Gmbh Process for the preparation of arachidonic acid and / or eicosapentaenoic acid
WO2008025068A1 (en) 2006-08-29 2008-03-06 Commonwealth Scientific And Industrial Research Organisation Synthesis of fatty acids
EP2054509A2 (en) 2006-10-06 2009-05-06 BASF Plant Science GmbH Processes for producing polyunsaturated fatty acids in transgenic organisms
US8916361B2 (en) 2006-11-17 2014-12-23 Abbott Laboratories Elongase gene and uses thereof
US7709239B2 (en) 2006-12-07 2010-05-04 E.I. Du Pont De Nemours And Company Mutant Δ8 desaturase genes engineered by targeted mutagenesis and their use in making polyunsaturated fatty acids
CN104152423A (en) 2007-04-03 2014-11-19 纳幕尔杜邦公司 Multizymes and their use in making polyunsaturated fatty acids
US7794701B2 (en) 2007-04-16 2010-09-14 E.I. Du Pont De Nemours And Company Δ-9 elongases and their use in making polyunsaturated fatty acids
US8957280B2 (en) 2007-05-03 2015-02-17 E. I. Du Pont De Nemours And Company Delta-5 desaturases and their use in making polyunsaturated fatty acids
US8119784B2 (en) 2008-04-02 2012-02-21 E. I. Du Pont De Nemours And Company Delta-4 desaturase and its use in making polyunsaturated fatty acids
CA2722275A1 (en) 2008-04-25 2009-10-29 Commonwealth Scientific And Industrial Research Organisation Polypeptides and methods for producing triacylglycerols comprising modified fatty acids
US8168858B2 (en) 2008-06-20 2012-05-01 E. I. Du Pont De Nemours And Company Delta-9 fatty acid elongase genes and their use in making polyunsaturated fatty acids
WO2010009500A1 (en) 2008-07-21 2010-01-28 Commonwealth Scientific And Industrial Research Organisation Improved vegetable oils and uses therefor
AU2009273754B2 (en) 2008-07-21 2016-07-14 Commonwealth Scientific And Industrial Research Organisation Improved cottonseed oil and uses
JP5757870B2 (en) 2008-08-26 2015-08-05 ビーエーエスエフ プラント サイエンス ゲーエムベーハー Nucleic acids encoding desaturases and modified vegetable oils
CN102186876B (en) 2008-10-14 2014-06-11 孟山都技术公司 Utilization of fatty acid desaturases from hemiselmis spp.
BR122021003835B1 (en) 2008-11-18 2022-02-08 Grains Research And Development Corporation ISOLATED AND/OR EXOGENOUS POLYNUCLEOTIDE THAT DOES NOT OCCUR IN A NATURAL WAY, VECTOR COMPRISING SUCH POLYNUCLEOTIDE AND PRODUCTION METHOD OF OIL CONTAINING UNSATURATED FATTY ACIDS
US8188335B2 (en) 2009-07-17 2012-05-29 Abbott Laboratories Δ9-elongase for production of polyunsaturated fatty acid-enriched oils
US9127288B2 (en) 2010-06-28 2015-09-08 Commonwealth Scientific And Industrial Research Organisation Methods of producing lipids
SG11201408362SA (en) 2012-06-15 2015-01-29 Commw Scient Ind Res Org Production of long chain polyunsaturated fatty acids in plant cells

Patent Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5504200A (en) 1983-04-15 1996-04-02 Mycogen Plant Science, Inc. Plant gene expression
US5463174A (en) 1986-05-29 1995-10-31 Calgene Inc. Transformation and foreign gene expression in Brassica species
US5608152A (en) 1986-07-31 1997-03-04 Calgene, Inc. Seed-specific transcriptional regulation
US5004863B1 (en) 1986-12-03 1992-12-08 Agracetus
US5004863A (en) 1986-12-03 1991-04-02 Agracetus Genetic engineering of cotton plants and lines
US5159135B1 (en) 1986-12-03 2000-10-24 Agracetus Genetic engineering of cotton plants and lines
US5159135A (en) 1986-12-03 1992-10-27 Agracetus Genetic engineering of cotton plants and lines
US5004863B2 (en) 1986-12-03 2000-10-17 Agracetus Genetic engineering of cotton plants and lines
US5416011A (en) 1988-07-22 1995-05-16 Monsanto Company Method for soybean transformation and regeneration
US5569834A (en) 1988-07-22 1996-10-29 Monsanto Company Method for soybean transformation and regeneration
WO1991013980A1 (en) 1990-03-16 1991-09-19 Calgene, Inc. Novel sequences preferentially expressed in early seed development and methods related thereto
US5518908A (en) 1991-09-23 1996-05-21 Monsanto Company Method of controlling insects
AU667939B2 (en) 1992-07-07 1996-04-18 Japan Tobacco Inc. Method of transforming monocotyledon
US20030163844A1 (en) * 1992-11-17 2003-08-28 Lightner Jonathan Edward Genes for microsomal delta-12 fatty acid desaturases and related enzymes from plants
US20030033633A1 (en) 1992-11-17 2003-02-13 Lightner Jonathan Edward Genes for microsomal delta-12 fatty acid desaturases and related enzymes from plants
AU6178194A (en) 1993-03-11 1994-09-26 National Research Council Of Canada Enhanced regeneration system for cereals
US5589617A (en) 1993-03-11 1996-12-31 National Research Council Of Canada Enhanced regeneration system
CA2092588A1 (en) 1993-03-26 1994-09-27 Narender S. Nehra Enhanced regeneration system for cereals
WO1997006269A1 (en) 1995-08-03 1997-02-20 Zeneca Limited Inducible herbicide resistance
US7109392B1 (en) * 1996-10-09 2006-09-19 Cargill, Incorporated Methods for increasing oleic acid content in seeds from transgenic plants containing a mutant delta 12 desaturase
WO1998045461A1 (en) 1997-04-09 1998-10-15 Rhone-Poulenc Agro An oleosin 5' regulatory region for the modification of plant seed lipid composition
US6432684B1 (en) 1997-04-11 2002-08-13 Abbott Laboratories Human desaturase gene and uses thereof
US6541257B2 (en) 1997-04-29 2003-04-01 The Regents Of The University Of California Method for preparing barley green regenerative tissue
WO1999014314A1 (en) 1997-09-12 1999-03-25 Commonwealth Scientific And Industrial Research Organisation Regulation of gene expression in plants
US6100447A (en) 1998-02-12 2000-08-08 Applied Phytologics, Inc. Method of barley transformation
WO1999049050A2 (en) * 1998-03-20 1999-09-30 E.I. Du Pont De Nemours And Company Limanthes oil genes
WO2001096363A1 (en) * 2000-06-12 2001-12-20 Boyce Thompson Institute For Plant Research Fatty acid desaturase gene and protein for modulating activation of defense signaling pathways in plants
US20070238648A1 (en) * 2000-09-26 2007-10-11 Brownlie Alison J Methods and compositions employing a novel stearoyl-CoA desaturase-hSCD5
US9710621B1 (en) 2011-11-02 2017-07-18 Google Inc. Platform for cloud application software

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
DAVID W STANLEY-SAMUELSON ET AL., ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY, vol. 9, 1 January 1988 (1988-01-01), pages 1 - 33
EIGENHEER ET AL., INSECT MOLECULAR BIOLOGY, vol. 11, no. 6, 2002, pages 533 - 542
EIGENHEER ET AL.: "Isolation and molecular characterization of Musca domestica delta-9 desaturase sequences", INSECT MOLECULAR BIOLOGY, vol. 11, no. 6, 2002, pages 533 - 542, XP008104613 *
HONG ET AL.: "Cis-3-Hexenal production in tobacco is stimulated by 16-carbon monounsaturated fatty acids", PHYTOCHEMISTRY, vol. 65, 2004, pages 159 - 168, XP004483375 *
KNIPPLE ET AL., GENETICS, vol. 162, 2002, pages 1737 - 1752
MORIMOTO ET AL., J. DAIRY SCI., vol. 88, 2005, pages 1142 - 1146
PARK HEE YUN ET AL., ENTOMOLOGICAL RESEARCH, vol. 35, no. 4, December 2005 (2005-12-01), pages 253 - 263
PASSORN S ET AL., BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 263, no. 1, 16 September 1999 (1999-09-16), pages 47 - 51
PEREIRA S L ET AL., BIOCHEMICAL JOURNAL, THE BIOCHEMICAL SOCIETY, vol. 378, no. 2, 1 January 2004 (2004-01-01), pages 665 - 671
PEYOU-NDI ET AL., ARCH OF BIOCHEM AND BIOPHYSICS, vol. 376, 2000, pages 399 - 408
RIDDERVOLD M.H. ET AL., INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 32, 2002, pages 1731 - 1740
RIDDERVOLD M.H. ET AL: "Biochemical and molecular characterization of house cricket (Acheta domesticus, Orthoptera: Gryllidae) delta9 desaturase", INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 32, 2002, pages 1731 - 1740, XP008104612 *
ROSENFIELD ET AL., INSECT BIOCHEM MOL BIOL, vol. 31, 2001, pages 949 - 964
See also references of EP2059588A4
WADA H ET AL., JOURNAL OF BACTERIOLOGY, vol. 175, no. 18, 1 September 1993 (1993-09-01), pages 6056 - 6058
WATTS AND BROWSE: "A palmitoyl-CoA-specific delta9 fatty acid desaturase from Caenorhabditis elegans", BIOCHEMISTRY AND BIOPHYSICS RESEARCH COMMUNICATIONS, vol. 272, 2000, pages 263 - 269, XP002191663 *
ZHOU ET AL., INSECT MOL BIOL, vol. 17, 2008, pages 667 - 676

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9963723B2 (en) 2004-04-22 2018-05-08 Commonwealth Scientific And Industrial Research Organisation Synthesis of long-chain polyunsaturated fatty acids by recombinant cells
US9458410B2 (en) 2004-04-22 2016-10-04 Commonwealth Scientific And Industrial Research Organisation Synthesis of long-chain polyunsaturated fatty acids by recombinant cell
US9453183B2 (en) 2004-04-22 2016-09-27 Commonwealth Scientific And Industrial Research Organisation Synthesis of long-chain polyunsaturated fatty acids by recombinant cell
US8853432B2 (en) 2004-04-22 2014-10-07 Commonwealth Scientific And Industrial Research Organisation Synthesis of long-chain polyunsaturated fatty acids by recombinant cell
US9951357B2 (en) 2004-04-22 2018-04-24 Commonweatlh Scientific And Industrial Research Organisation Synthesis of long-chain polyunsaturated fatty acids by recombinant cell
US8530724B2 (en) 2006-07-14 2013-09-10 Commonwealth Scientific And Industrial Research Organisation Altering the fatty acid composition of rice
US9351507B2 (en) 2006-07-14 2016-05-31 Commonwealth Scientific And Industrial Research Organisation Method of preparing food using rice oil
US10260021B2 (en) 2006-07-14 2019-04-16 Commonwealth Scientific And Industrial Research Organisation Rice plants and methods of producing rice grain
US10513717B2 (en) 2006-08-29 2019-12-24 Commonwealth Scientific And Industrial Research Organisation Synthesis of fatty acids
US9057075B2 (en) 2008-07-21 2015-06-16 Commonwealth Scientific And Industrial Research Organisation Cottonseed oil and uses
US8921652B2 (en) 2008-07-21 2014-12-30 Commonwealth Scientific And Industrial Research Organisation Vegetable oils and uses therefor
US8716555B2 (en) 2008-07-21 2014-05-06 Commonwealth Scientific And Industrial Research Organisation Cottonseed oil and uses
US9938486B2 (en) 2008-11-18 2018-04-10 Commonwealth Scientific And Industrial Research Organisation Enzymes and methods for producing omega-3 fatty acids
US10006065B2 (en) 2010-03-02 2018-06-26 Massachusetts Institute Of Technology Microbial engineering for the production of fatty acids and fatty acid derivatives
US8765404B2 (en) 2010-03-02 2014-07-01 Massachusetts Institute Of Technology Microbial engineering for the production of fatty acids and fatty acid derivatives
US9127288B2 (en) 2010-06-28 2015-09-08 Commonwealth Scientific And Industrial Research Organisation Methods of producing lipids
US10925293B2 (en) 2010-06-28 2021-02-23 Commonwealth Scientific And Industrial Research Organisation Methods of producing lipids
WO2012003545A1 (en) * 2010-07-09 2012-01-12 Commonwealth Scientific And Industrial Research Organisation Acetylenation of fatty acids
US10246641B2 (en) 2011-12-27 2019-04-02 The Commonwealth Scientific And Industrial Research Organisation Processes for producing hydrocarbon products
US8809026B2 (en) 2011-12-27 2014-08-19 Commonwealth Scientific And Industrial Research Organisation Processes for producing lipids
US9512438B2 (en) 2011-12-27 2016-12-06 Commonwealth Scientific And Industrial Research Organisation Processes for producing hydrocarbon products
US8735111B2 (en) 2011-12-27 2014-05-27 Commonwealth Scientific And Industrial Research Organisation Processes for producing hydrocarbon products
US9499829B2 (en) 2011-12-27 2016-11-22 Commonwealth Scientific And Industrial Research Organisation Processes for producing lipids
US10246718B2 (en) 2011-12-27 2019-04-02 The Commonwealth Scientific And Industrial Research Organisation Processes for producing lipids
US9061992B2 (en) 2011-12-27 2015-06-23 Commonwealth Scientific And Industrial Research Organisation Processes for producing hydrocarbon products
US11639507B2 (en) 2011-12-27 2023-05-02 Commonwealth Scientific And Industrial Research Organisation Processes for producing lipids
US10323209B2 (en) 2012-04-25 2019-06-18 Commonwealth Scientific And Industrial Research Organisation High oleic acid oils
US11124737B2 (en) 2012-04-25 2021-09-21 Commonwealth Scientific Andn Industrial Research Organisation High oleic acid oils
US8946460B2 (en) 2012-06-15 2015-02-03 Commonwealth Scientific And Industrial Research Organisation Process for producing polyunsaturated fatty acids in an esterified form
US10335386B2 (en) 2012-06-15 2019-07-02 Commonwealth Scientific And Industrial Research Organisation Lipid comprising polyunsaturated fatty acids
US9556102B2 (en) 2012-06-15 2017-01-31 Commonwealth Scientific And Industrial Research Organisation Process for producing ethyl esters of polyunsaturated fatty acids
US9550718B2 (en) 2012-06-15 2017-01-24 Commonwealth Scientific And Industrial Research Organisation Lipid comprising polyunsaturated fatty acids
US9932289B2 (en) 2012-06-15 2018-04-03 Commonwealth Scientific And Industrial Research Ogranisation Process for producing ethyl esters of polyunsaturated fatty acids
US10800729B2 (en) 2013-12-18 2020-10-13 Commonwealth Scientific And Industrial Research Organisation Lipid comprising long chain polyunsaturated fatty acids
US9718759B2 (en) 2013-12-18 2017-08-01 Commonwealth Scientific And Industrial Research Organisation Lipid comprising docosapentaenoic acid
US9725399B2 (en) 2013-12-18 2017-08-08 Commonwealth Scientific And Industrial Research Organisation Lipid comprising long chain polyunsaturated fatty acids
US10190073B2 (en) 2013-12-18 2019-01-29 Commonwealth Scientific And Industrial Research Organisation Lipid comprising long chain polyunsaturated fatty acids
US11623911B2 (en) 2013-12-18 2023-04-11 Commonwealth Scientific And Industrial Research Organisation Lipid comprising docosapentaenoic acid
US10125084B2 (en) 2013-12-18 2018-11-13 Commonwealth Scientific And Industrial Research Organisation Lipid comprising docosapentaenoic acid
US10793507B2 (en) 2014-06-27 2020-10-06 Commonwealth Scientific And Industrial Research Organisation Lipid compositions comprising triacylglycerol with long-chain polyunsaturated fatty acids at the SN-2 position
US10005713B2 (en) 2014-06-27 2018-06-26 Commonwealth Scientific And Industrial Research Organisation Lipid compositions comprising triacylglycerol with long-chain polyunsaturated fatty acids at the sn-2 position
US10472587B2 (en) 2014-07-07 2019-11-12 Commonwealth Scientific And Industrial Research Organisation Processes for producing industrial products from plant lipids
US11365369B2 (en) 2014-07-07 2022-06-21 Commonwealth Scientific And Industrial Research Organisation Processes for producing industrial products from plant lipids
US11814600B2 (en) 2014-07-07 2023-11-14 Nuseed Global Innnovation Ltd. Process for producing industrial products from plant lipids
US11859193B2 (en) 2016-09-02 2024-01-02 Nuseed Global Innovation Ltd. Plants with modified traits

Also Published As

Publication number Publication date
EP2059588A1 (en) 2009-05-20
EP2059588A4 (en) 2010-07-28
CN101578363A (en) 2009-11-11
US10513717B2 (en) 2019-12-24
RU2009111266A (en) 2010-10-10
CA2661697A1 (en) 2008-03-06
BRPI0716075A2 (en) 2013-08-06
AU2007291937A1 (en) 2008-03-06
US20150045567A1 (en) 2015-02-12
AU2007291937B2 (en) 2014-05-15
US8816106B2 (en) 2014-08-26
US20110190521A1 (en) 2011-08-04

Similar Documents

Publication Publication Date Title
US10513717B2 (en) Synthesis of fatty acids
US20240150780A1 (en) Enzymes and methods for producing omega-3 fatty acids
KR102527795B1 (en) Lipid comprising docosapentaenoic acid
KR102386838B1 (en) Lipid comprising long chain polyunsaturated fatty acids
US20110218348A1 (en) Polypeptides and methods for producing triacylglycerols comprising modified fatty acids
US20110126325A1 (en) Recombinant cells and methods for hydroxylating fatty acids
JP2021107552A (en) Lipid comprising docosapentaenoic acid
AU2013204270B8 (en) Enzymes and methods for producing omega-3 fatty acids
WO2012003545A1 (en) Acetylenation of fatty acids
AU2015224521B2 (en) Enzymes and methods for producing omega-3 fatty acids

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780040245.5

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07784864

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2661697

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007291937

Country of ref document: AU

Ref document number: 575809

Country of ref document: NZ

REEP Request for entry into the european phase

Ref document number: 2007784864

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007784864

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2009111266

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2007291937

Country of ref document: AU

Date of ref document: 20070829

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12310645

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0716075

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20090302