NZ621037B2 - Genes and proteins for alkanoyl-coa synthesis - Google Patents

Genes and proteins for alkanoyl-coa synthesis Download PDF

Info

Publication number
NZ621037B2
NZ621037B2 NZ621037A NZ62103712A NZ621037B2 NZ 621037 B2 NZ621037 B2 NZ 621037B2 NZ 621037 A NZ621037 A NZ 621037A NZ 62103712 A NZ62103712 A NZ 62103712A NZ 621037 B2 NZ621037 B2 NZ 621037B2
Authority
NZ
New Zealand
Prior art keywords
nucleic acid
coa
acid molecule
cell
cannabinoid
Prior art date
Application number
NZ621037A
Other versions
NZ621037A (en
Inventor
Jonathan E Page
Jason M Stout
Original Assignee
National Research Council Of Canada
University Of Saskatchewan
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 National Research Council Of Canada, University Of Saskatchewan filed Critical National Research Council Of Canada
Priority claimed from PCT/CA2012/000656 external-priority patent/WO2013006953A1/en
Publication of NZ621037A publication Critical patent/NZ621037A/en
Publication of NZ621037B2 publication Critical patent/NZ621037B2/en

Links

Classifications

    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/52Genes encoding for enzymes or proenzymes
    • 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)
    • 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
    • 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
    • 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
    • C12N9/10Transferases (2.)
    • C12N9/1025Acyltransferases (2.3)
    • C12N9/1029Acyltransferases (2.3) transferring groups other than amino-acyl groups (2.3.1)
    • 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
    • C12N9/90Isomerases (5.)
    • 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
    • C12N9/93Ligases (6)
    • 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
    • C12P17/00Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms
    • C12P17/02Oxygen as only ring hetero atoms
    • C12P17/06Oxygen as only ring hetero atoms containing a six-membered hetero ring, e.g. fluorescein
    • 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
    • C12P7/40Preparation of oxygen-containing organic compounds containing a carboxyl group including Peroxycarboxylic acids
    • C12P7/42Hydroxy-carboxylic acids
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/52Improvements relating to the production of bulk chemicals using catalysts, e.g. selective catalysts

Abstract

Discloses polypeptides having alkanoyl-CoA activity, nucleic acids encoding the same, and methods relating to altering levels or synthesising cannabinoid compounds in an organism, cell or tissue.

Description

GENES AND PROTEINS FOR ALKANOYL-COA SYNTHESIS Cross-reference to Related Applications This application claims the benefit of United States Provisional Application Serial Number USSN 61/507,331 filed July 13, 2011, the entire contents of which is herein incorporated by reference.
Field of the Invention The present invention relates to nucleic acid molecules and proteins involved in the synthesis of alkanoyl-CoA thioesters, and to uses of the nucleic acid molecules and proteins for engineering cannabinoid biosynthesis in plants, micro-organisms or in cell- free systems and for creating cannabis plants with enhanced or reduced cannabinoid content.
Background of the Invention Cannabis sativa L. (cannabis, hemp, marijuana) is one of the oldest and most versatile domesticated plants, which today finds use as source of medicinal, food, cosmetic and industrial products. It is also well known for its use as an illicit drug owing to its content of psychoactive cannabinoids (e.g. -tetrahydrocannabinol, -THC) .
Cannabinoids and other drugs that act through mammalian cannabinoid receptors are being explored for the treatment of diverse conditions such as chronic pain, multiple sclerosis and epilepsy.
Cannabinoids have their biosynthetic origins in both polyketide and terpenoid metabolism and are termed terpenophenolics or prenylated polyketides (P age J., Nagel J. (2006) Biosynthesis of terpenophenolics in hop and cannabis. In JT Romeo, ed, Integrative Plant Biochemistry, Vol. 40. Elsevier, Oxford, pp 179-210.). Cannabinoid biosynthesis occurs primarily in glandular trichomes that cover female flowers at a high density. Cannabinoids are formed by a three-step biosynthetic process:polyketide formation, aromatic prenylation and cyclization (see Figure 1).
The first enzymatic step in cannabinoid biosynthesis is the formation of olivetolic acid by a polyketide synthase enzyme that catalyzes the condensation of hexanoyl- coenzyme A (CoA) with three molecules of malonyl-CoA. The major cannabinoids, including -tetrahydrocannabinolic acid and cannabidiolic acid, are formed from the precursor hexanoyl-CoA, which is a medium chain fatty acyl-CoA (see Figure 1). Other cannabinoids with variant side-chains are formed from aliphatic-CoAs of different lengths (e.g. -tetrahydrocannabivarinic acid is formed from an n-butyryl-CoA primer).
Hexanoyl-CoA and other acyl-CoA thioesters in plants are synthesized by acyl- activating enzymes (AAEs, also called acyl-CoA synthetases) that catalyze the activation of carboxylic acid substrates using ATP. These enzymes act on a variety of carboxylate acids including short-, medium-, long- and very long-chain fatty acids, jasmonate precursors, phenylpropanoid-derived acids (e.g. cinnamic acid) and other organic acids such as malonate, acetate and citrate. Very few medium-chain acyl CoA synthetases have been previously identified in nature. In plants, three enzymes from A. thaliana, AAE7, At4g05160 and At5g63380 have been shown to form hexanoyl-CoA from hexanoate (Schneider K et al. (2005) A new type of peroxisomal acyl-coenzyme A synthetase from Arabidopsis thaliana has the catalytic capacity to activate biosynthetic precursors of jasmonic acid. The Journal of Biological Chemistry 280:13962-72; Shockey JM, Fulda MS, Browse J (2003) Arabidopsis contains a large superfamily of acyl- activating enzymes. Phylogenetic and biochemical analysis reveals a new class of acyl- coenzyme a synthetases. Plant Physiology 132:1065-76.) Acyl-CoA synthetases from Pseudomonas spp. have been shown to act on medium-chain fatty acids such as hexanoate (Fernandez-Valverde M, Reglero A, Martinez-Blanco H, Luengo JM (1993) Purification of Pseudomonas putida acyl coenzyme A ligase active with a range of aliphatic and aromatic substrates. Applied Environmental Microbiology 59:1149-1154.) Cannabinoids are valuable natural products. Genes encoding enzymes involved in cannabinoid biosynthesis will be useful in metabolic engineering of cannabis to produce plants that contain very low levels, or zero levels, of THCA and other cannabinoids via targeted mutagenesis (e.g. TILLING) or other gene knockout techniques. Such genes may also prove useful for creation, via marker-assisted selection, of specific cannabis varieties for the production of cannabinoid-based pharmaceuticals, or for reconstituting cannabinoid biosynthesis in heterologous organisms such as bacteria or yeast, or for producing cannabinoids in cell-free systems that utilize recombinant proteins.
Genes encoding enzymes of cannabinoid biosynthesis can also be useful in synthesis of cannabinoid analogs and synthesis of analogs of cannabinoid precursors.
Cannabinoid analogs have been previously synthesized and may be useful as pharmaceutical products.
There remains a need in the art to identify enzymes, and nucleotide sequences encoding such enzymes, that are involved in the synthesis of aromatic polyketides.
Summary of the Invention Two novel genes from cannabis have now been found which encode previously unknown alkanoyl-CoA synthetases. These two new alkanoyl Co-A synthetases are referred to herein as Cannabis sativa hexanoyl-CoA synthetase 1 (CsHCS1) and Cannabis sativa hexanoyl-CoA synthetase 2 (CsHCS2) .
Thus, in a first aspect of the invention, there is provided an isolated or purified nucleic acid molecule comprising a nucleotide sequence having at least 75% sequence identity to SEQ ID NO:1, or a codon degenerate sequence thereof.
In a second aspect of the invention, there is provided an isolated or purified nucleic acid molecule comprising a nucleotide sequence having at least 75% sequence identity to SEQ ID NO:3, or a codon degenerate sequence thereof.
In a third aspect of the invention, there is provided an isolated or purified polypeptide comprising an amino acid sequence having at least 85% sequence identity to SEQ ID NO:2, or a conservatively substituted amino acid sequence thereof.
In a fourth aspect of the invention, there is provided an isolated or purified polypeptide comprising an amino acid sequence having at least 85% sequence identity to SEQ ID NO:4, or a conservatively substituted amino acid sequence thereof.
In a fifth aspect of the invention, there is provided a vector, construct or expression system comprising a nucleic acid molecule of the invention.
In a sixth aspect of the invention, there is provided a host cell transformed with a nucleic acid molecule of the invention.
In a seventh aspect of the invention, there is provided a process of synthesizing an alkanoyl-CoA in presence of an enzyme of the invention.
In an eighth aspect of the invention, there is provided a process of altering levels of cannabinoid compounds in an organism, cell or tissue comprising using a nucleic acid molecule of the present invention, or a part thereof, to silence in the organism, cell or tissue a gene that encodes an enzyme that catalyzes synthesis of an alkanoyl-CoA.
In an ninth aspect of the invention, there is provided a process of altering levels of cannabinoid compounds in an organism, cell or tissue comprising mutating genes in the organism, cell or tissue, and using a nucleic acid molecule of the present invention to select for organisms, cells or tissues containing mutants or variants of a gene that encodes an enzyme that catalyzes synthesis of an alkanoyl-CoA.
In a tenth aspect of the invention, there is provided a process of altering levels of cannabinoid compounds in an organism, cell or tissue comprising expressing or over- expressing a nucleic acid molecule of the invention in the organism, cell or tissue in comparison to a similar variety of organism, cell or tissue grown under similar conditions but without the expressing or over-expressing of the nucleic acid molecule.
In an eleventh aspect of the invention, there is provided a process of altering levels of cannabinoid compounds in an organism, cell or tissue comprising expressing or over-expressing a nucleic acid molecule encoding a polypeptide of the invention in the organism, cell or tissue in comparison to a similar variety of organism, cell or tissue grown under similar conditions but without the expressing or over-expressing of the nucleic acid molecule.
In a twelfth aspect of the invention, there is provided a process of synthesizing a naturally-occurring cannabinoid compound or a non-naturally occurring analog of a cannabinoid compound in an organism, cell or tissue comprising expressing a nucleic acid molecule of the invention in the organism, cell or tissue in the presence of a carboxylic acid and CoA.
In a thirteenth aspect of the present invention, there is provided a process of synthesizing an alkanoyl-CoA in an in vitro cell-free reaction, said process comprising: reacting a carboxylic acid with coenzyme A presence of an enzyme of the invention.
Polypeptides that are enzymes catalyzing the synthesis of alkanoyl-CoA, and nucleotide sequences encoding such enzymes, have now been identified and characterized. The nucleotide sequences may be used to create, through breeding, selection or genetic engineering, cannabis plants that overproduce or under-produce cannabinoid compounds, analogs of cannabinoid compounds or mixtures thereof. These nucleotide sequences may also be used, alone or in combination with genes encoding other steps in cannabinoid synthesis pathways, to engineer cannabinoid biosynthesis in other plants or in microorganisms (e.g. yeast, bacteria, fungi) or other prokaryotic or eukaryotic organisms or in cell-free systems. In addition, blocking or reducing the expression of these genes in cannabis could be used to block cannabinoid biosynthesis and thereby reduce production of cannabinoids.
Further features of the invention will be described or will become apparent in the course of the following detailed description.
Brief Description of the Drawings In order that the invention may be more clearly understood, embodiments thereof will now be described in detail by way of example, with reference to the accompanying drawings, in which: Figure 1 depicts a proposed pathway leading to the main cannabinoid types in Cannabis sativa. Abbreviations:THCA synthase is -tetrahydrocannabinolic acid synthase; CBDA synthase is cannabidiolic acid synthase; CBCA synthase is cannabichromenic acid synthase.
Figures 2A-2F depict liquid chromatography mass spectrometry/mass spectrometry (LC-MS/MS) analysis of the enzymatic activity of Cannabis sativa hexanoyl- CoA synthases. Each of Figures 2A-2F show ion abundance (m /z 866>359) on the vertical axis and time (minutes) on the horizontal axis. Figure 2A and 2B depict the retention time of an authentic hexanoyl-CoA standard. Figure 2C depicts an assay of CsHCS1 protein, CoA, MgCl , sodium hexanoate, ATP, and HEPES buffer, in which hexanoyl-CoA was produced and detected. Figure 2D depicts an assay of CsHCS2 protein, CoA, MgCl , sodium hexanoate, ATP, and HEPES buffer, in which hexanoyl-CoA was produced. Figure 2E depicts an assay in which CsHCS1 protein had been previously inactivated by boiling at 95°C for 15 minutes, CoA, sodium hexanoate, ATP, and HEPES buffer, in which no hexanoyl-CoA was produced. Fig 2F depicts an assay in which CsHCS2 protein had been previously inactivated by boiling at 95°C for 15 minutes, CoA, sodium hexanoate, ATP, and HEPES buffer, in which no hexanoyl-CoA was produced.
Figure 3 depicts two graphs illustrating carboxylic acid substrates utilized by the enzymes of the invention. Figure 3A depicts carboxylic acid substrates utilized by CsHCS1. Figure 3B depicts carboxylic acid substrates utilized by CsHCS2.
Figure 4 depicts a high performance liquid chromatography analysis of the products produced by a coupled enzymatic assay consisting of the Cannabis sativa hexanoyl-CoA synthetase CsHCS2, malonyl-CoA synthetase ( MCS), Cannabis sativa olivetol synthase/polyketide synthase, and Cannabis sativa olivetolic acid synthase.
Eluted compounds were detected by absorbance at 263 nm and identified both by having the same retention times as isolated standards, and by their mass using a single quadrapole mass detector. The detection of olivetol and olivetolic acid indicates that CsHCS2 is capable of providing sufficient hexanoyl-CoA substrate for the synthesis of olivetolic acid. Assays lacking CsHCS2, CoA, or hexanoate did not produce any polyketide products. HTAL = hexanoyltriacetic lactone, PDAL = pentyldiacetic lactone, OA = olivetolic acid, OL = olivetol.
Figure 5 depicts a graph showing olivetolic acid production in yeast cells engineered to produce olivetolic acid by using CsHCS1 and CsHCS2 to synthesize hexanoyl-CoA, and a fusion of the cannabis olivetol synthase/polyketide synthase (PKS) and olivetolic acid synthase (O AS) to form olivetolic acid.
Figure 6 depicts qRT-PCR analysis of CsHCS1, CsHCS2 and CBDA Synthase expression in different tissues of the hemp cultivar F inola. Gene expression values relative to actin were plotted as fold differences compared to leaves, with leaf expression assigned a value of 1. Insets depict gene expression in female flowers with and without trichomes, with values also indicated as fold differences compared to leaves. R, roots; S, stems; L, leaves; FF+, female flowers with trichomes; FF-, female flowers with trichomes removed by the Beadbeater method; T, trichomes; MF, male flower. Values are mean ± SD, n=3.
Description of Preferred Embodiments A trichome-specific cDNA library deom cannabis was sequenced to produce 9157 express sequence tags ( ESTs) that assembled into 4113 unique sequences ( 1227 contigs, 2886 singletons). Unigenes were annotated by comparison to the UniProt protein database using the online search and comparison tool called blastx. Cannabis acyl-activating enzyme proteins were identified by utilizing Arabidopsis acyl-activating enzyme sequences to query the assembled cannabis ESTs using the online search and comparison tool called tblastn. Eleven acyl-activating enzymes were identified and named according to their transcript abundance in the cDNA library. CsHCS1 was the most abundant acyl-activating enzyme based on transcript levels (42 ESTs); CsHCS2 had lower abundance (5 ESTs) Based on its high transcript levels in trichomes and the localization of CsHCS1 to the cytoplasm, it is likely that this enzyme is the acyl-activating enzyme involved in supplying hexanoyl-CoA to the cannabinoid pathway. CsHCS2, which is localized to the peroxisome, is probably not involved in cannabinoid formation.
However, its kinetic properties make it a useful enzyme for synthesizing hexanoyl-CoA in heterologous hosts or in cell-free systems.
The sequence of the CsHCS1 gene is as follows: Cannabis sativa CsHCS1 2163 bp (SEQ ID NO:1) ATGGGTAAGAATTACAAGTCCCTGGACTCTGTTGTGGCCTCTGACTTCATAGCCCTA GGTATCACCTCTGAAGTTGCTGAGACACTCCATGGTAGACTGGCCGAGATCGTGTG TAATTATGGCGCTGCCACTCCCCAAACATGGATCAATATTGCCAACCATATTCTGTCG CCTGACCTCCCCTTCTCCCTGCACCAGATGCTCTTCTATGGTTGCTATAAAGACTTTG GACCTGCCCCTCCTGCTTGGATACCCGACCCGGAGAAAGTAAAGTCCACCAATCTG GGCGCACTTTTGGAGAAGCGAGGAAAAGAGTTTTTGGGAGTCAAGTATAAGGATCC CATTTCAAGCTTTTCTCATTTCCAAGAATTTTCTGTAAGAAACCCTGAGGTGTATTGG AGAACAGTACTAATGGATGAGATGAAGATAAGTTTTTCAAAGGATCCAGAATGTATAT TGCGTAGAGATGATATTAATAATCCAGGGGGTAGTGAATGGCTTCCAGGAGGTTATC TTAACTCAGCAAAGAATTGCTTGAATGTAAATAGTAACAAGAAATTGAATGATACAAT GATTGTATGGCGTGATGAAGGAAATGATGATTTGCCTCTAAACAAATTGACACTTGAC CAATTGCGTAAACGTGTTTGGTTAGTTGGTTATGCACTTGAAGAAATGGGTTTGGAG AAGGGTTGTGCAATTGCAATTGATATGCCAATGCATGTGGATGCTGTGGTTATCTAT CTAGCTATTGTTCTTGCGGGATATGTAGTTGTTTCTATTGCTGATAGTTTTTCTGCTC CTGAAATATCAACAAGACTTCGACTATCAAAAGCAAAAGCCATTTTTACACAGGATCA TATTATTCGTGGGAAGAAGCGTATTCCCTTATACAGTAGAGTTGTGGAAGCCAAGTC TCCCATGGCCATTGTTATTCCTTGTAGTGGCTCTAATATTGGTGCAGAATTGCGTGAT GGCGATATTTCTTGGGATTACTTTCTAGAAAGAGCAAAAGAGTTTAAAAATTGTGAAT TTACTGCTAGAGAACAACCAGTTGATGCCTATACAAACATCCTCTTCTCATCTGGAAC AACAGGGGAGCCAAAGGCAATTCCATGGACTCAAGCAACTCCTTTAAAAGCAGCTG CAGATGGGTGGAGCCATTTGGACATTAGGAAAGGTGATGTCATTGTTTGGCCCACTA ATCTTGGTTGGATGATGGGTCCTTGGCTGGTCTATGCTTCACTCCTTAATGGGGCTT CTATTGCCTTGTATAATGGATCACCACTTGTTTCTGGCTTTGCCAAATTTGTGCAGGA TGCTAAAGTAACAATGCTAGGTGTGGTCCCTAGTATTGTTCGATCATGGAAAAGTAC CAATTGTGTTAGTGGCTATGATTGGTCCACCATCCGTTGCTTTTCCTCTTCTGGTGAA GCATCTAATGTAGATGAATACCTATGGTTGATGGGGAGAGCAAACTACAAGCCTGTT ATCGAAATGTGTGGTGGCACAGAAATTGGTGGTGCATTTTCTGCTGGCTCTTTCTTA CAAGCTCAATCATTATCTTCATTTAGTTCACAATGTATGGGTTGCACTTTATACATACT TGACAAGAATGGTTATCCAATGCCTAAAAACAAACCAGGAATTGGTGAATTAGCGCT TGGTCCAGTCATGTTTGGAGCATCGAAGACTCTGTTGAATGGTAATCACCATGATGT TTATTTTAAGGGAATGCCTACATTGAATGGAGAGGTTTTAAGGAGGCATGGGGACAT TTTTGAGCTTACATCTAATGGTTATTATCATGCACATGGTCGTGCAGATGATACAATG AATATTGGAGGCATCAAGATTAGTTCCATAGAGATTGAACGAGTTTGTAATGAAGTTG ATGACAGAGTTTTCGAGACAACTGCTATTGGAGTGCCACCTTTGGGCGGTGGACCT GAGCAATTAGTAATTTTCTTTGTATTAAAAGATTCAAATGATACAACTATTGACTTAAA 40 TCAATTGAGGTTATCTTTCAACTTGGGTTTACAGAAGAAACTAAATCCTCTGTTCAAG GTCACTCGTGTTGTGCCTCTTTCATCACTTCCGAGAACAGCAACCAACAAGATCATG AGAAGGGTTTTGCGCCAGCAATTTTCTCACTTTGAATGA The sequence of the CsHCS2 gene is as follows: Cannabis sativa CsHCS2 1547 bp (SEQ ID NO:3) ATGGAGAAATCTTTTTCAGAAACTCATCTTCATACCCACAAAAGCCAGCTCTCATTGA TTCCGAAACCAACCAAATACTCTCCTTTTCCCACTTCAAATCTACGGTTATCAAGGTC TCCCATGGCTTTCTCAATCTGGGTATCAAGAAAAACGACGTCGTTCTCATCTACGCC CCTAATTCTATCCACTTCCCTGTTTGTTTCCTGGGAATTATAGCCTCTGGAGCCATTG CCACTACCTCAAATCCTCTCTACACAGTTTCCGAGCTTTCCAAACAGGTCAAGGATTC CAATCCCAAACTCATTATCACCGTTCCTCAACTCTTGGAAAAAGTAAAGGGTTTCAAT CTCCCCACGATTCTAATTGGTCCTGATTCTGAACAAGAATCTTCTAGTGATAAAGTAA TGACCTTTAACGATTTGGTCAACTTAGGTGGGTCGTCTGGCTCAGAATTTCCAATTGT TGATGATTTTAAGCAGAGTGACACTGCTGCGCTATTGTACTCATCTGGCACAACGGG AATGAGTAAAGGTGTGGTTTTGACTCACAAAAACTTCATTGCCTCTTCTTTAATGGTG ACAATGGAGCAAGACCTAGTTGGAGAGATGGATAATGTGTTTCTATGCTTTTTGCCA ATGTTTCATGTATTTGGTTTGGCTATCATCACCTATGCTCAGTTGCAGAGAGGAAACA CTGTTATTTCAATGGCGAGATTTGACCTTGAGAAGATGTTAAAAGATGTGGAAAAGTA TAAAGTTACCCATTTGTGGGTTGTGCCTCCTGTGATACTGGCTCTGAGTAAGAACAG TATGGTGAAGAAGTTTAATCTTTCTTCTATAAAGTATATTGGCTCCGGTGCAGCTCCT TTGGGCAAAGATTTAATGGAGGAGTGCTCTAAGGTTGTTCCTTATGGTATTGTTGCTC AGGGATATGGTATGACAGAAACTTGTGGGATTGTATCCATGGAGGATATAAGAGGAG GTAAACGAAATAGTGGTTCAGCTGGAATGCTGGCATCTGGAGTAGAAGCCCAGATA GTTAGTGTAGATACACTGAAGCCCTTACCTCCTAATCAATTGGGGGAGATATGGGTG AAGGGGCCTAATATGATGCAAGGTTACTTCAATAACCCACAGGCAACCAAGTTGACT ATAGATAAGAAAGGTTGGGTACATACTGGTGATCTTGGATATTTTGATGAAGATGGA CATCTTTATGTTGTTGACCGTATAAAAGAGCTCATCAAATATAAAGGATTTCAGGTTG CTCCTGCTGAGCTTGAAGGATTGCTTGTTTCTCACCCTGAAATACTCGATGCTGTTGT GATTCCATTTCCTGACGCTGAAGCGGGTGAAGTCCCAGTTGCTTATGTTGTGCGCTC TCCCAACAGTTCATTAACCGAAAATGATGTGAAGAAATTTATCGCGGGCCAGGTTGC ATCTTTCAAAAGATTGAGAAAAGTAACATTTATAAACAGTGTCCCGAAATCTGCTTCG GGGAAAATCCTCAGAAGAGAACTCATTCAGAAAGTACGCTCCAACATGTGA CsHCS1 and CsHCS2 were PCR amplified as described in Example 1 and the alkanoyl-CoA synthetase or CoA-ligase activity was measured as described in Example 2. As is shown in Figure 2, CsHCS1 and CsHCS2 catalyze the production of alkanoyl- CoA from a carboxylic acid and CoA.
Some embodiments of the present invention relate to an isolated or purified nucleic acid molecule having SEQ ID NO:1 or having at least 75%, at least 76%, least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 40 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to SEQ ID NO:1.
Some embodiments of the present invention relate to an isolated or purified nucleic acid molecule having SEQ ID NO:3 or having at least 75%, at least 76%, least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to SEQ ID NO:3.
Further included are nucleic acid molecules that hybridize to the above disclosed nucleic acid sequences. Hybridization conditions may be stringent in that hybridization will occur if there is at least a 90%, 95% or 97% sequence identity with the nucleic acid molecule that encodes the enzyme of the present invention. The stringent conditions may include those used for known Southern hybridizations such as, for example, incubation overnight at 42 C in a solution having 50% formamide, 5x SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate ( pH 7.6), 5x Denhardt's solution, 10% dextran sulfate, and 20 micrograms/milliliter denatured, sheared salmon sperm DNA, following by washing the hybridization support in 0.1x SSC at about 65°C. Other known hybridization conditions are well known and are described in Sambrook et al., Molecular Cloning:A Laboratory Manual, Third Edition, Cold Spring Harbor, N.Y. (2001) .
As will be appreciated by the skilled practitioner, slight changes in nucleic acid sequence do not necessarily alter the amino acid sequence of the encoded polypeptide.
It will be appreciated by persons skilled in the art that changes in the identities of nucleotides in a specific gene sequence that change the amino acid sequence of the encoded polypeptide may result in reduced or enhanced effectiveness of the genes and that, in some applications (e.g. anti-sense, co suppression, or RNAi) , partial sequences often work as effectively as full length versions. The ways in which the nucleotide sequence can be varied or shortened are well known to persons skilled in the art, as are ways of testing the effectiveness of the altered genes. In certain embodiments, effectiveness may easily be tested by, for example, conventional gas chromatography.
All such variations of the genes are therefore included as part of the present disclosure.
As will be appreciated by one of skill in the art, the length of the nucleic acid molecule described above will depend on the intended use. For example, if the intended use is as a primer or probe, for example for PCR amplification or for screening a library, the length of the nucleic acid molecule will be less than the full length sequence, for example, 15-50 nucleotides. In these embodiments, the primers or probes may be substantially identical to a highly conserved region of the nucleic acid sequence or may be substantially identical to either the 5' or 3' end of the DNA sequence. In some cases, these primers or probes may use universal bases in some positions so as to be 'substantially identical' but still provide flexibility in sequence recognition. It is of note that suitable primer and probe hybridization conditions are well known in the art.
The present invention also includes the enzyme CsHCS1. The amino acid sequence of CsHCS1 (SEQ ID NO:2) is: MGKNYKSLDSVVASDFIALGITSEVAETLHGRLAEIVCNYGAATPQTWINIANHILSPDLPF SLHQMLFYGCYKDFGPAPPAWIPDPEKVKSTNLGALLEKRGKEFLGVKYKDPISSFSHF QEFSVRNPEVYWRTVLMDEMKISFSKDPECILRRDDINNPGGSEWLPGGYLNSAKNCL NVNSNKKLNDTMIVWRDEGNDDLPLNKLTLDQLRKRVWLVGYALEEMGLEKGCAIAIDM PMHVDAVVIYLAIVLAGYVVVSIADSFSAPEISTRLRLSKAKAIFTQDHIIRGKKRIPLYSRV VEAKSPMAIVIPCSGSNIGAELRDGDISWDYFLERAKEFKNCEFTAREQPVDAYTNILFSS GTTGEPKAIPWTQATPLKAAADGWSHLDIRKGDVIVWPTNLGWMMGPWLVYASLLNGA SIALYNGSPLVSGFAKFVQDAKVTMLGVVPSIVRSWKSTNCVSGYDWSTIRCFSSSGEA SNVDEYLWLMGRANYKPVIEMCGGTEIGGAFSAGSFLQAQSLSSFSSQCMGCTLYILDK NGYPMPKNKPGIGELALGPVMFGASKTLLNGNHHDVYFKGMPTLNGEVLRRHGDIFELT SNGYYHAHGRADDTMNIGGIKISSIEIERVCNEVDDRVFETTAIGVPPLGGGPEQLVIFFV LKDSNDTTIDLNQLRLSFNLGLQKKLNPLFKVTRVVPLSSLPRTATNKIMRRVLRQFSHFE Some embodiments relate to an isolated or purified polypeptide having SEQ ID NO. 2 or having at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to the amino acid sequence as set forth in SEQ ID NO:2.
The present invention also includes the enzyme CsHCS2. The amino acid sequence of CsHCS2 (SEQ ID NO:4) is: MEKSGYGRDGIYRSLRPPLHLPNNNNLSMVSFLFRNSSSYPQKPALIDSETNQILSFSHF KSTVIKVSHGFLNLGIKKNDVVLIYAPNSIHFPVCFLGIIASGAIATTSNPLYTVSELSKQVK DSNPKLIITVPQLLEKVKGFNLPTILIGPDSEQESSSDKVMTFNDLVNLGGSSGSEFPIVD DFKQSDTAALLYSSGTTGMSKGVVLTHKNFIASSLMVTMEQDLVGEMDNVFLCFLPMFH VFGLAIITYAQLQRGNTVISMARFDLEKMLKDVEKYKVTHLWVVPPVILALSKNSMVKKFN LSSIKYIGSGAAPLGKDLMEECSKVVPYGIVAQGYGMTETCGIVSMEDIRGGKRNSGSA GMLASGVEAQIVSVDTLKPLPPNQLGEIWVKGPNMMQGYFNNPQATKLTIDKKGWVHT GDLGYFDEDGHLYVVDRIKELIKYKGFQVAPAELEGLLVSHPEILDAVVIPFPDAEAGEVP VAYVVRSPNSSLTENDVKKFIAGQVASFKRLRKVTFINSVPKSASGKILRRELIQKVRSNM Some embodiments relate to an isolated or purified polypeptide having SEQ ID NO. 4 or having at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to the amino acid sequence as set forth in SEQ ID NO:4.
Some embodiments relate to a vector, construct or expression system containing 40 an isolated or purified polynucleotide having the sequence of SEQ ID NO:1 or SEQ ID NO:3, or least 75%, at least 76%, least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to SEQ ID NO:1 or SEQ ID NO:3. As well, there is provided a method for preparing a vector, construct or expression system including such a sequence, or a part thereof, for introduction of the sequence or partial sequence in a sense or anti-sense orientation, or a complement thereof, into a cell.
In some embodiments, the isolated and/or purified nucleic acid molecules, or vectors, constructs or expression systems comprising these isolated and/or purified nucleic acid molecules, may be used to create transgenic organisms or cells of organisms that produce polypeptides which catalyze the synthesis of aromatic polyketides.
Therefore, one embodiment relates to transgenic organisms, cells or germ tissues of the organism comprising an isolated and/or purified nucleic acid molecule having SEQ ID NO:1 or SEQ ID NO:3 or having least 75%, at least 76%, least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to SEQ ID NO:1 or SEQ ID NO:3.
Preferably, the organism is a plant, microorganism or insect. Plants are preferably of the genus Cannabis, for example Cannabis sativa L., Cannabis indica Lam. and Cannabis ruderalis Janisch. Especially preferred is Cannabis sativa. Microorganisms are preferably bacteria (e.g. Escherichia coli) or yeast (e.g. Saccharomyces cerevisiae).
Insect is preferably Spodoptera frugiperda.
Organisms, cells and germ tissues of this embodiment may have altered levels of cannabinoid compounds. With reference to Figure 1, it will be appreciated by one skilled in the art that expression or over-expression of the nucleic acid molecules of the invention will result in expression or over-expression of the enzyme that catalyzes the synthesis of hexanoyl-CoA which, in combination with other enzymes, may result in the production or increased production of cannabinoid compounds such as cannabigerolic acid (C BGA), - tetrahydrocannabinolic acid (T HCA), cannabidiolic acid (CBDA) , cannabichromenic acid (CBCA), -tetrahydrocannabinol ( THC), cannabidiol (CBD), cannabichromene (CBC), etc. Similarly, depending on the substrate used, expression or over-expression of the nucleic acid molecules of the invention resulting in expression or over-expression of the enzyme that catalyzes the synthesis of hexanoyl-CoA may result in the production or increased production of analogs of cannabinoid compounds, or analogs of precursors of such compounds.
Silencing of the gene in the organism, cell or tissue will result in under-expression of the enzyme which may result in accumulation of precursors such as hexanoic acid (six carbons), octanoic acid (eight carbons), nonanoic acid (nine carbons), valeric acid (f ive carbons), heptanoic acid (seven carbons) or other carboxylic acids, and/or reduction of cannabinoids such as THCA (the precursor of THC) or CBDA (the precursor of CBD).
The present invention includes a process of altering levels of cannabinoid compounds in an organism, cell or tissue by expressing or over-expressing an exogenous enzyme of the invention in the organism, cell or tissue, in comparison to a similar variety of organism, cell or tissue grown under similar conditions but without an exogenous enzyme of the invention being expressed or over-expressed.
Expression or over-expression of the nucleic acid molecules of the invention may be done in combination with expression or over-expression of one or more other nucleic acids that encode one or more enzymes in a cannabinoid biosynthetic pathway. Some examples of other nucleic acids include those which encode:a type III polyketide synthase, a polyketide cyclase, an aromatic prenyltransferase and a cannabinoid-forming oxidocylase. Specific examples of these enzymes include olivetol synthase/polyketide synthase, olivetolic acid synthase, a geranylpyrophosphate:olivetolate geranyltransferase, a -tetrahydrocannabinolic acid synthase, a cannabidiolic acid synthase or a cannabichromenic acid synthase. Synthesis of alkanoyl-CoA in the presence of an enzyme polypeptide of the present invention may be accomplished in vivo or in vitro. As previously mentioned, such syntheses in vivo may be accomplished by expressing or over-expressing the nucleic acid molecule of the invention in an organism, cell or tissue.
Synthesis of alkanoyl-CoA in vitro can take place in a cell-free system. As part of an in vitro cell-free system, the carboxylic acid and an enzyme of the present invention may be mixed together in a suitable reaction vessel to effect the reaction.
In vitro, the polypeptides of the present invention may be used in combination with other enzymes to effect a complete synthesis of a cannabinoid compound from a precursor. For example, such other enzymes may be implicated in a cannabinoid biosynthetic pathway as described in Figure 1 ( su ch as olivetol synthase/PKS , olivetolic acid synthase, aromatic prenyltransferase, THCA synthase, CBDA synthase, CBCA synthase).
The polypeptides of the present invention may be used, in vivo or in vitro, to synthesize analogs of cannabinoid compounds which are not naturally occurring in the host species. Such analogs can be produced using carboxylic acid compounds other than those used to produce natural cannabinoid compounds in plants. For example, acetic acid, butyric acid, octanoic acid, decanoic acid, lauric acid, myristic acid, palmitic acid; branched chain acids such isovaleric acid; and hydroxycinnamic acids such a cinnamic acid.
Terms: In order to facilitate review of the various embodiments of the disclosure, the following explanations of specific terms are provided: Alkanoyl-CoA: An alkanoyl-CoA is an aliphatic carbonyl compound having a coenzyme A moiety bonded to the carbon atom of the carbonyl group through a sulfide bridge. Preferred alkanoyl-CoA compounds comprise from 2 to 10 carbon atoms in the aliphatic carbonyl part of the compound. More preferably, the alkanoyl-CoA is CoA-S- C(O)- (CH ) -CH , where n is an integer from 0 to 8. Some examples of alkanoyl-CoA 2 n 3 compounds are acetyl-CoA, butyryl-CoA, hexanoyl-CoA and octanoyl-CoA. Use of acetyl-CoA provides a methyl side chain to the resulting aromatic polyketide; use of butyryl-CoA provides a propyl side chain; and use of hexanoyl-CoA provides a pentyl side chain. Hexanoyl-CoA is especially preferred. Cannabinoids with shorter side-chains exist in cannabis (e.g. tetrahydrocannabivarinic acid having a propyl side-chain instead of the pentyl side-chain of THCA).
Codon degeneracy: It will be appreciated that this disclosure embraces the degeneracy of codon usage as would be understood by one of ordinary skill in the art and as illustrated in Table 1.
Table 1 Codon Degeneracies Amino Acid Codons Ala/A GCT, GCC, GCA, GCG Arg/R CGT, CGC, CGA, CGG, AGA, AGG Asn/N AAT, AAC Asp/D GAT, GAC Cys/C TGT, UGC Gln/Q CAA, CAG Glu/E GAA, GAG Gly/G GGT, GGC, GGA, GGG His/H CAT, CAC Ile/I ATT, ATC, ATA Leu/L TTA, TTG, CTT, CTC, CTA, CTG Lys/K AAA, AAG Met/M ATG Phe/F TTT, TTC Pro/P CCT, CCC, CCA, CCG Ser/S TCT, TCC, TCA, TCG, AGT, AGC Thr/T ACT, ACC, ACA, ACG Trp/W TGG Tyr/Y TAT, TAC Val/V GTT, GTC, GTA, GTG START ATG STOP TAG, TGA, TAA Complementary nucleotide sequence: Complementary nucleotide sequence of a sequence is understood as meaning any nucleic acid molecule whose nucleotides are complementary to those of a sequence disclosed herein, and whose orientation is reversed (anti-parallel sequence).
Conservative substitutions: It will be understood by one skilled in the art that conservative substitutions may be made in the amino acid sequence of a polypeptide without disrupting the three-dimensional structure or function of the polypeptide.
Accordingly, the present invention includes polypeptides comprising conservatively substituted CsHCS1 and CsHCS2. Conservative substitutions are accomplished by the skilled artisan by substituting amino acids with similar hydrophobicity, polarity, and R- chain length for one another. Additionally, by comparing aligned sequences of homologous proteins from different species, conservative substitutions may be identified by locating amino acid residues that have been mutated between species without altering the basic functions of the encoded proteins. Table 2 provides an exemplary list of conservative substitutions.
Table 2 Conservative Substitutions Type of Amino Acid Substitutable Amino Acids Hydrophilic Ala, Pro, Gly, Glu, Asp, Gln, Asn, Ser, Thr Sulphydryl Cys Aliphatic Val, Ile, Leu, Met Basic Lys, Arg, His Aromatic Phe, Tyr, Trp Degree or percentage of sequence homology: The term "degree or percentage of sequence homology'' refers to degree or percentage of sequence identity between two sequences after optimal alignment.
Homologous isolated and/or purified sequence: Homologous isolated and/or purified sequence is unde rstood to mean an isolated and/or purified sequence having a percentage identity with the bases of a nucleotide sequence, or the amino acids of a polypeptide sequence, of at least about 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.6%, or 99.7%. This percentage is purely statistical, and it is possible to distribute the differences between the two nucleotide or amino acid sequences at random and over the whole of their length. Sequence identity can be determined, for example, by computer programs designed to perform single and multiple sequence alignments.
Increasing, decreasing, modulating, altering or the like: As will be appreciated by one of skill in the art, such terms refer to comparison to a similar variety or strain grown under similar conditions but without the modification resulting in the increase, decrease, modulation or alteration. In some cases, this may be an untransformed control, a mock transformed control, or a vector-transformed control.
Isolated: As will be appreciated by one of skill in the art, isolated refers to polypeptides or nucleic acids that have been isolated from their native environment.
Nucleotide, polynucleotide, or nucleic acid sequence: Nucleotide, polynucleotide, or nucleic acid sequence will be understood as meaning both double -stranded or single- stranded in the monomeric and dimeric (so-called in tandem) forms and the transcription products thereof.
Sequence identity: Two amino acid or nucleotide sequences are said to be "identical'' if the sequence of amino acids or nucleotides in the two sequences is the same when aligned for maximum correspondence as described below. Percentage of sequence identity (or degree of identity) is determined by comparing two optimally aligned sequences over a comparison window, where the portion of the peptide or polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical amino acid residue or nucleic acid base occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
Optimal alignment of sequences for comparison may be conducted by the local homology algorithm of Smith and Waterman, Ad. App. Math 2:482 (1 981), by the homology alignment algorithm of Needleman and Wunsch, J. Mol. Biol. 48:443 (1970), by the search for similarity method of Pearson and Lipman, Proc. Natl. Acad. Sci. ( U.S.A.) 85:2444 (1988), by computerized implementation of these algorithms (G AP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (G CG), 575 Science Dr., Madison, Wis.), or by visual inspection.
The definition of sequence identity given above is the definition that would be used by one of skill in the art. The definition by itself does not need the help of any algorithm, said algorithms being helpful only to achieve the optimal alignments of sequences, rather than the calculation of sequence identity.
From the definition given above, it follows that there is a well defined and only one value for the sequence identity between two compared sequences which value corresponds to the value obtained for the best or optimal alignment.
Stringent hybridization: Hybridization under conditions of stringency with a nucleotide sequence is understood as meaning a hybridization under conditions of temperature and ionic strength chosen in such a way that they allow the maintenance of the hybridization between two fragments of complementary nucleic acid molecules.
Homologs of the novel genes described herein obtained from other organisms, for example plants, may be obtained by screening appropriate libraries that include the homologs, wherein the screening is performed with the nucleotide sequence of the specific genes of the invention, or portions or probes thereof, or identified by sequence homology search using sequence alignment search programs such as BLAST or FASTA.
Nucleic acid isolation and cloning is well established. Similarly, an isolated gene may be inserted into a vector and transformed into a cell by conventional techniques which are known to those of skill in the art. Nucleic acid molecules may be transformed into an organism. As known in the art, there are a number of ways by which genes, vectors, constructs and expression systems can be introduced into organisms, and a combination of transformation and tissue culture techniques have been successfully integrated into effective strategies for creating transgenic organisms. These methods, which can be used in the invention, have been described elsewhere ( P otrykus I (1991) Gene transfer to plants:Assessment of published approaches and results. Annu. Rev.
Plant Physiol. Plant Mol. Biol. 42:205-225; Vasil I K (1994) Molecular improvement of cereals. Plant Mol. Biol. 25:925-937. Walden R, Wingender R (1995) Gene-transfer and plant regeneration techniques. Trends in Biotechnology 13:324-331; Songstad DD, Somers DA, Griesbach RJ ( 1995) Advances in alternative DNA delivery techniques. Plant Cell Tissue Organ Cult. 40:1-15), and are well known to persons skilled in the art.
Suitable vectors are well known to those skilled in the art and are described in general technical references such as Pouwels et al., Cloning Vectors. A Laboratory Manual, Elsevier, Amsterdam (1986) . Particularly suitable vectors include the Ti plasmid vectors. For example, one skilled in the art will certainly be aware that, in addition to Agrobacterium mediated transformation of Arabidopsis by vacuum infiltration (Bechtold N, Ellis J, Pelletier G (1993) In planta Agrobacterium-mediated gene transfer by infiltration of adult Arabidopsis thaliana plants. C R Acad Sci Paris, Sciences de la vie/Life sciences 316:1194-1199.) or wound inoculation (Katavic V, Haughn GW, Reed D, Martin M, Kunst L (1994) In planta transformation of Arabidopsis thaliana. Mol. Gen. Genet. 245:363- 370.) , it is equally possible to transform other plant species, using Agrobacterium Ti- plasmid mediated transformation (e.g., hypocotyl (DeBlock M, DeBrouwer D, Tenning P (1989) Transformation of Brassica napus and Brassica oleracea using Agrobacterium tumefaciens and the expression of the bar and neo genes in the transgenic plants. Plant Physiol. 91:694-701) or cotyledonary petiole (Moloney MM, Walker JM, Sharma KK (1989) High efficiency transformation of Brassica napus using Agrobacterium vectors.
Plant Cell Rep. 8:238-242.) wound infection, particle bombardment/biolistic methods (Sanford JC, Klein TM, Wolf ED, Allen N (1987) Delivery of substances into cells and tissues using a particle bombardment process. J. Part. Sci. Technol. 5:27-37.) or polyethylene glycol-assisted, protoplast transformation methods ( Rhodes CA, Pierce DA, Mettler IJ, Mascarenhas D, Detmer JJ (1988) Genetically transformed maize plants from protoplasts. Science 240:204-207).
As will also be apparent to persons skilled in the art, and as described elsewhere (Meyer P ( 1995) Understanding and controlling transgene expression. Trends in Biotechnology 13:332-337; Datla R, Anderson JW, Selvaraj G (1997) Plant promoters for transgene expression. Biotechnology Annual Review 3:269-296.), it is possible to utilize promoters operatively linked to the nucleic acid molecule to direct any intended up- or down-regulation of transgene expression using unregulated (i.e. constitutive) promoters (e.g., those based on CaMV35S) , or by using promoters which can target gene expression to particular cells, tissues (e.g., napin promoter for expression of transgenes in developing seed cotyledons), organs (e .g., roots), to a particular developmental stage, or in response to a particular external stimulus (e.g., heat shock).
Promoters for use in the invention may be inducible, constitutive, or tissue-specific or have various combinations of such characteristics. Useful promoters include, but are not limited to constitutive promoters such as carnation etched ring virus promoter (CERV), cauliflower mosaic virus (CaMV) 35S promoter, or more particularly the double enhanced cauliflower mosaic virus promoter, comprising two CaMV 35S promoters in tandem (referred to as a "Double 35S" promoter). It may be desirable to use a tissue- specific or developmentally regulated promoter instead of a constitutive promoter in certain circumstances. A tissue-specific promoter allows for over-expression in certain tissues without affecting expression in other tissues.
The promoter and termination regulatory regions will be functional in the host cell and may be heterologous (that is, not naturally occurring) or homologous (derived from the host species) to the cell and the gene.
The termination regulatory region may be derived from the 3' region of the gene from which the promoter was obtained or from another gene. Suitable termination regions which may be used are well known in the art and include Agrobacterium tumefaciens nopaline synthase terminator (T nos), A. tumefaciens mannopine synthase terminator (T mas) and the CaMV 35S terminator (T 35S). Particularly preferred termination regions for use in the present invention include the pea ribulose bisphosphate carboxylase small subunit termination region ( TrbcS) or the Tnos termination region.
Gene constructs for use in the invention may suitably be screened for activity by, for example, transformation into a host plant via Agrobacterium and screening for altered cannabinoid levels.
The nucleic acid molecules of the invention, or fragments thereof, may be used to block cannabinoid biosynthesis in organisms that naturally produce cannabinoid compounds. Silencing using a nucleic acid molecule of the invention may be accomplished in a number of ways generally known in the art, for example, RNA interference ( RNAi) techniques, artificial microRNA techniques, virus-induced gene silencing (VIGS) techniques, antisense techniques, sense co-suppression techniques and targeted mutagenesis techniques.
RNAi techniques involve stable transformation using RNA interference (RNAi) plasmid constructs (Helliwell CA, Waterhouse PM (2005) Constructs and methods for hairpin RNA-mediated gene silencing in plants. Methods Enzymology 392:24-35). Such plasmids are composed of a fragment of the target gene to be silenced in an inverted repeat structure. The inverted repeats are separated by a spacer, often an intron. The RNAi construct driven by a suitable promoter, for example, the Cauliflower mosaic virus (CaMV) 35S promoter, is integrated into the plant genome and subsequent transcription of the transgene leads to an RNA molecule that folds back on itself to form a double- stranded hairpin RNA. This double-stranded RNA structure is recognized by the plant and cut into small RNAs (about 21 nucleotides long) called small interfering RNAs (siRNAs). The siRNAs associate with a protein complex (RISC) which goes on to direct degradation of the mRNA for the target gene.
Artificial microRNA (amiRNA) techniques exploit the microRNA (miRNA) pathway that functions to silence endogenous genes in plants and other eukaryotes (Schwab R, Ossowski S, Riester M, Warthmann N, Weigel D ( 2006) Highly specific gene silencing by artificial microRNAs in Arabidopsis. Plant Cell 18:1121-33; Alvarez JP, Pekker I, Goldshmidt A, Blum E, Amsellem Z, Eshed Y (2006) Endogenous and synthetic microRNAs stimulate simultaneous, efficient, and localized regulation of multiple targets in diverse species. Plant Cell 18:1134-51). In this method, 21 nucleotide long fragments of the gene to be silenced are introduced into a pre-miRNA gene to form a pre-amiRNA construct. The pre-amiRNA construct is transferred into the organism genome using transformation methods which would be apparent to one skilled in the art. After transcription of the pre-amiRNA, processing yields amiRNAs that target genes which share nucleotide identity with the 21 nucleotide amiRNA sequence.
In RNAi silencing techniques, two factors can influence the choice of length of the fragment. The shorter the fragment the less frequently effective silencing will be achieved, but very long hairpins increase the chance of recombination in bacterial host strains. The effectiveness of silencing also appears to be gene dependent and could reflect accessibility of target mRNA or the relative abundances of the target mRNA and the hairpin RNA in cells in which the gene is active. A fragment length of between 100 and 800 bp, preferably between 300 and 600 bp, is generally suitable to maximize the efficiency of silencing obtained. The other consideration is the part of the gene to be targeted. 5 UTR, coding region, and 3 UTR fragments can be used with equally good results. As the mechanism of silencing depends on sequence homology, there is potential for cross-silencing of related mRNA sequences. Where this is not desirable, a region with low sequence similarity to other sequences, such as a 5 or 3 UTR, should be chosen. The rule for avoiding cross-homology silencing appears to be to use sequences that do not have blocks of sequence identity of over 20 bases between the construct and the non-target gene sequences. Many of these same principles apply to selection of target regions for designing amiRNAs.
Virus-induced gene silencing (VIGS) techniques are a variation of RNAi techniques that exploits the endogenous antiviral defenses of plants. Infection of plants with recombinant VIGS viruses containing fragments of host DNA leads to post- transcriptional gene silencing for the target gene. In one embodiment, a tobacco rattle virus ( TRV) based VIGS system can be used with the nucleotide sequences of the present invention.
Antisense techniques involve introducing into a plant an antisense oligonucleotide that will bind to the messenger RNA (mRNA) produced by the gene of interest. The "antisense" oligonucleotide has a base sequence complementary to the gene's messenger RNA (mRNA), which is called the "sense" sequence. Activity of the sense segment of the mRNA is blocked by the anti-sense mRNA segment, thereby effectively inactivating gene expression. Application of antisense to gene silencing in plants is described in more detail by Stam M, de Bruin R, van Blokland R, van der Hoorn RA, Mol JN, Kooter JM (2000) Distinct features of post-transcriptional gene silencing by antisense transgenes in single copy and inverted T-DNA repeat loci. Plant J. 21:27-42.
Sense co-suppression techniques involve introducing a highly expressed sense transgene into a plant resulting in reduced expression of both the transgene and the endogenous gene (D epicker A, Montagu MV ( 1997) Post-transcriptional gene silencing in plants. Curr Opin Cell Biol. 9:373-82). The effect depends on sequence identity between transgene and endogenous gene.
Targeted mutagenesis techniques, for example TILLING (T argeting Induced Local Lesions IN Genomes) and delete -a-gene using fast -neutron bombardment, may be used to knockout gene function in an organism ( Henikoff S, Till BJ, Comai L (2004) TILLING. Traditional mutagenesis meets functional genomics. Plant Physiol 135:630-6; Li X, Lassner M, Zhang Y. (2002) Deleteagene:a fast neutron deletion mutagenesis-based gene knockout system for plants. Comp Funct Genomics. 3:158-60). TILLING involves treating germplasm or individual cells with a mutagen to cause point mutations that are then discovered in genes of interest using a sensitive method for single-nucleotide mutation detection. Detection of desired mutations (e.g. mutations resulting in the inactivation of the gene product of interest) may be accomplished, for example, by PCR methods. For example, oligonucleotide primers derived from the gene of interest may be prepared and PCR may be used to amplify regions of the gene of interest from organisms in the mutagenized population. Amplified mutant genes may be annealed to wild-type genes to find mismatches between the mutant genes and wild-type genes. Detected differences may be traced back to the organism which had the mutant gene thereby revealing which mutagenized organism will have the desired expression (e.g. silencing of the gene of interest) . These organisms may then be selectively bred to produce a population having the desired expression. TILLING can provide an allelic series that includes missense and knockout mutations, which exhibit reduced expression of the targeted gene. TILLING is touted as a possible approach to gene knockout that does not involve introduction of transgenes, and therefore may be more acceptable to consumers.
Fast-neutron bombardment induces mutations, i.e. deletions, in organism genomes that can also be detected using PCR in a manner similar to TILLING.
It will be understood by one of skill in the art that the processes of the invention can also be carried out in a cell-free environment in the presence of one or more carboxylic acids.
Embodiments of the invention are susceptible to various modifications and alternative forms in addition to the specific examples included herein. Thus, embodiments of the invention are not limited to the particular forms disclosed.
Examples: Example 1:Amplification and Cloning of CsHCS1 and CsHCS2 CsHCS1 and CsHCS2 were PCR amplified from cDNA plasmid clones using the primers listed in Table 3 and Phusion polymerase (Finnzymes). PCR products were purified and cloned into the pCR8/GW/TOPO entry vector ( Invitrogen) . After transformation into E. coli TOP10 cells ( I nvitrogen) , individual clones were verified by sequencing. The CsHCS1 and CsHCS2 genes were recombined into the pHIS8/GW destination vector using LR recombinase (I nvitrogen) . The LR reaction products were transformed into TOP10 cells and verified by sequencing.
Table 3: Oligonucleotides Name Sequence ( 5 -3) CsHCS1 forward (SEQ ID NO:5) ATGGGTAAGAATTACAAGTCCCT CsHCS1 reverse (SEQ ID NO:6) GAGCTCTCATTCAAAGTGAGAAAATTGCTG CsHCS2 forward (SEQ ID NO:7) ATGGAGAAATCTGGGTATGGAAG CsHCS2 reverse (SEQ ID NO:8) TCACATGTTGGAGCGTACTTTC MCS forward (SEQ ID NO:9) ATGAGCAACCATCTTTTCGACG MCS reverse (SEQ ID NO:10) TTACGTCCTGGTATAAAGATCGGC pHIS8/GW-CsHCS1 and pHIS8/GW-CsHCS2 were transformed into E. coli Rosetta 2 cells ( Merck). Individual colonies were used to inoculate small-scale cultures of liquid LB medium containing chloramphenicol and kanamycin, which were used to inoculate 500 mL of liquid LB medium without antibiotics. After growth to OD of 0.6, expression was induced by the addition of IPTG to 0.2 µM. The CsHCS1 expressing cultures were then grown at 12°C with shaking for 24 h, whereas the CsHCS2 cultures were grown at 37°C for 16 h. Different temperatures were used because it was observed that CsHCS1 did not produce soluble protein at the higher temperature.
Cells were harvested by centrifugation and resuspended in 10 mL His-tag lysis buffer ( 50 mM Tris-HCl pH 7, 500 mM NaCl, 2.5 mM imidazole, 10% v/v glycerol, 10 mM -mercaptoethanol, 1% v/v Tween 20, and 750 µg/mL lysozyme) . The resuspended cells were incubated on ice for 1 h then lysed by sonication. After centrifugation for 20 min at 12,000 g at 4°C, the soluble protein fraction was added to 160 µL of Talon resin ( Clontech) that had previously been washed with His-tag wash buffer ( HWB; 50 mM Tris- HCl pH 7, 500 mM NaCl, 2.5 mM imidazole, 10% glycerol, 10 mM -mercaptoethanol).
The samples were incubated with gentle rocking at 4°C, after which the resin was isolated by centrifugation (700 g for 5 min). The resin was resuspended in HWB buffer and washed with gentle rocking at 4°C then centrifuged. The wash step was then repeated twice and the resuspended resin loaded onto a chromatography column and allowed to drain. After washing the resin with 10 mL of HWB buffer, the His-tagged proteins were eluted by the addition of 2.5 mL of His-tag elution buffer (50 mM Tris-HCl pH 7, 500 mM NaCl, 250 mM imidazole, 10% v/v glycerol, 10 mM -mercaptoethanol). The eluates were buffer exchanged into storage buffer (50 mM HEPES pH 9, 10% v/v glycerol, 2 mM MgCl , and 2 mM dithiothreitol) using PD10 columns (Amersham Biosciences). The purity of the isolated proteins was verified by SDS-PAGE, and the protein concentration was determined by Bradford assay.
Example 2:Analysis of Hexanoyl-CoA Synthetase Activity Hexanoyl-CoA synthetase activity was measured by incubating 0.1 µg of enzyme in a 20 µL reaction mixture containing 50 mM HEPES pH 9, 8 mM ATP, 10 mM MgCl , 0.5 mM CoA, and 4 mM sodium hexanoate. The reactions were incubated for 10 min at 40°C, terminated with 2 µL of 1 N HCl and stored on ice until analysis.
The reaction mixtures were diluted 1:100 with water and subsequently separated using a Waters Acquity UPLC system fitted with an Acquity UPLC BEH C18 column (1.7 µm particle size, 2.1 x 50 mm column), and analyzed by MS/MS using a Micromass Quattro Ultima triple-quadrupole mass spectrometer. The solvent system used was buffer A:5 mM TEA and 3 mM acetic acid in water, and buffer B:5 mM TEA and 3 mM acetic acid in 95:5 methanol:water. The flow program is shown in Table 4. The mass spectrometer settings were:ESI positive mode, collision energy 27 V, cone 135 V, scanning for 866>359 transitions.
Table 4: Flow Program for Liquid Chromatography As shown in Figure 2, CsHCS1 and CsHCS2 catalyzed the formation of hexanoyl- CoA from hexanoate and CoA. Figures 2A and 2B show the elution of authentic hexanoyl-CoA standard. Figure 2C shows a complete assay comprising CsHCS1, 50 mM HEPES pH 9, 8 mM ATP, 10 mM MgCl , 0.5 mM CoA, and 4 mM sodium hexanoate. A compound with the same mass transitions and elution time as the authentic hexanoyl- CoA standard can be seen at 9.25 minutes. Figure 2D shows a complete assay comprising of CsHCS2, 50 mM HEPES pH 9, 8 mM ATP, 10 mM MgCl , 0.5 mM CoA, and 4 mM sodium hexanoate. A compound with the same mass transitions and elution time as the authentic hexanoyl-CoA standard can be seen at 9.25 minutes. Figures 2E and 2F show negative controls with inactivated (boiled) CsHCS1 and CsHCS2 enzymes.
As can be seen in Figures 2E and 2F, these assays showed no hexanoyl-CoA synthesis.
Both CsHCS1 and CsHCS2 exhibited temperature and pH optima of 40°C and pH 9, respectively. In testing a range of divalent cations, CsHCS1 optimally used Mg and to 2+ 2+ 2+ a lesser extent Mn and Co . CsHCS2 activity was highest using Co , but was also 2+ 2+ 2+ observed to be high with Mg , Mn , and to a lesser extent Ca . The biological relevance 2+ 2+ of the high activity with Co is not clear and Mg was used for all further assays.
With hexanoate, CsHCS1 had a K of 6.1 ± 1.0 mM, a V of 15.6 ± 1.7 pKat and m max a k of 4.5 sec . CsHCS2 had a K of 320 nM, a V of 1.7 pKat, and a k of 57.6 cat m max cat sec .
Example 3:Testing with Different Carboxylic Acids To test the range of carboxylic acids that CsHCS1 and CsHCS2 can activate, enzyme assays were performed with a broad range of carboxylic acids and limiting ATP.
The assay conditions used were similar to Schneider K et al. (2005). A new type of peroxisomal acyl-coenzyme A synthetase from Arabidopsis thaliana has the catalytic capacity to activate biosynthetic precursors of jasmonic acid. The Journal of Biological Chemistry, 280:13962-72. Briefly, purified HCS enzyme (1 µg) was incubated with 500 µM carboxylic acid substrate and 100 µM CoA in an assay containing 100 mM HEPES pH 9, 250 µM MgCl , 50 µM ATP and 1 mM DTT. All carboxylic acid substrates were dissolved in 2% v/v Triton X-100, leading to a final concentration 0.05% Triton X-100 in the assay. After reacting for 3 h at 29°C, 10 µL aliquots of the reactions were transferred to 96-well plates for a luciferin/luciferase based measurement of unconsumed ATP. The plates were analyzed with a 1420 Multilabel counter (PerkinElmer). To each well, 90 µL of a solution containing 100 mM Tris pH 7.8, 1 mM MgCl , 2.3 µg luciferin, and 0.5 µg of luciferase was injected, and after shaking for 2 seconds the luminescence was measured for 15 seconds without a filter in place. Lower readings, compared to the reactions with no carboxylic acid substrate, indicate a higher amount of enzymatic activity and therefore substrate utilization. The results are shown in Figure 3, wherein error bars represent the percent error of the ratio, n=3.
As is shown in Figure 3A, CsHCS1 was observed to utilize hexanoate ( six carbons), octanoate ( eight carbons), and nonanoate (nine carbons), and to a lesser extent valerate (f ive carbons) and heptanoate (seven carbons), as substrates. In contrast, as is shown in Figure 3B, CsHCS2 exhibited greater promiscuity, and is able to utilize a broad range of substrates, ranging from propanoate (C3) to arachidoate (C20), and a number of phenylpropanoids (cinnamate, ferulate, and to a lesser extent p-coumarate).
In a separate experiment, the kinetic properties of CsHCS1 and CsHCS3 were more accurately measured for CoA and representative short (butanoate), medium ( hexanoate and decanoate), and long-chain (palmitate) fatty acids ( T able 5). High CoA concentrations inhibited CsHCS1. Using a non-linear regression substrate inhibition model, the K of CsHCS1 was estimated to be 5.101 ± 1.8 mM. CoA did not inhibit CsHCS2 at the concentrations tested. Decanoic acid inhibited CsHCS2 (K = 120.8 ± 47.9 µM) and slightly inhibited CsHCS1 in concentrations above 4 mM (K not measured) .
These data show the same trends as the kinetic data presented above.
Table 5:Kinetic properties of CsHCS1 and CsHCS2.
Substrate CsHCS1 CsHCS2 K V ( pKat) K V ( pKat) k (s ) m max m max cat CoA 0.26 ± 0.05 µM - 0.16 ± 0.01 µM - - butanoate >10 mM - >10 mM - - hexanoate 3.7 ± 0.7 mM 6.8 ± 0.7 261 ± 37 µM 1.8 ± 0.05 57.6 decanoate 1.7 ± 0.2 mM 1.8 ± 0.7 16.1 ± 5.8 µM 1.6 ± 0.1 10.0 palmitate n.d. - 1.3 ± 0.5 µM 0.4 ± 0.01 2.4 not determined due to lack of catalytic activity Example 4:Synthesis of Olivetolic Acid using CsHCS2 CsHCS2 was used for the chemoenzymatic synthesis of the aromatic polyketide olivetolic acid. Olivetolic acid is the first committed precursor for cannabinoid biosynthesis. This in vitro synthesis made use of four recombinant enzymes:CsHCS2, malonyl-CoA synthetase (MCS) from Rhizobium leguminosarum, olivetol synthase/polyketide synthase from cannabis, and olivetolic acid synthase from cannabis.
Malonyl-CoA synthetase (MCS) was amplified from genomic DNA of Rhizobium leguminosarum with the primers MCS forward and MCS reverse (see Table 3). The PCR product was cloned into pCR8/GW/TOPO vector (Invitrogen) recombined into pHIS8/GW vector. After verification by sequencing, the plasmid was transformed into the E. coli Rosetta II (DE3). Recombinant MCS was expressed and purified as described for the CsHCS enzymes.
The cloning, expression and purification of olivetol synthase/ polyketide synthase and olivetolic acid synthase were done as follows. For expression in E. coli cells, the open reading frames of polyketide synthase/olivetol synthase and olivetolic acid synthase were amplified by PCR, cloned into pHIS8/GW for polyketide synthase/olivetol synthase or pET100 ( I nvitrogen) for olivetolic acid synthase and transformed into E. coli BL21 (DE3) (Invitrogen) . Cloning was verified by sequencing.
Olivetolic acid synthase was expressed in 200 mL terrific broth culture while polyketide synthase/olivetol synthase grown in a 1 L culture. Both cultures were incubated at 30°C/150 rpm shaking, induced with 0.5 M IPTG and grown overnight. The cultures were centrifuged at 16,000 g for 20 min, and the pellets lysed by treatment with lysozyme and sonication. The cleared lysates were mixed with Talon resin (200 L for olivetolic acid synthase, 1 mL for polyketide synthase/olivetol synthase; Clontech), washed with 5 mL of His-tag Wash Buffer (50 mM Tris-HCl (pH 7), 150 mM NaCl, 20 mM imidazole, 10 mM -mercaptoethanol) and the recombinant proteins eluted using His-tag Elution Buffer (2 0 mM Tris HCl (pH 7), 150 mM NaCl, 100 mM imidazole, 10 mM - mercaptoethanol). The eluate was concentrated using a YM10 concentrator and the buffer exchanged to Storage Buffer (20 mM HEPES (pH 7.5), 25 mM NaCl, 10% glycerol, mM DTT). The final protein solutions were quantified by using an RC/DC protein assay kit (Bio-Rad) which found protein concentrations of 0.5 mg/mL (olivetolic acid synthase) and 5.6 mg/mL (polyketide synthase/olivetol synthase). SDS-PAGE gel analysis confirmed the purity of both proteins.
The ability to couple hexanoyl-CoA synthesis with aromatic polyketide synthesis using inexpensive reagents was tested by performing enzyme assays consisting of 4 mM hexanoate, 8 mM malonate, 0.4 mM CoA, 0.4 mM ATP, 5 mM MgCl , 2 mM DTT, 20 mM HEPES pH 7.5, 0.3 µg CsHCS2, 12 µg MCS, 8 µg olivetol synthase/PKS and 10 µg OAS. The reaction was incubated at room temperature for 16 h, acidified and extracted in ethyl acetate. The polar fraction was recovered, evaporated to dryness and resuspended in 50 µL of methanol. An aliquot ( 5 µL) was analyzed by LCMS, and products were identified by their retention times and masses (see Figure 4).
Example 5:Synthesis of Olivetolic Acid in Yeast using CsHCS1 and CsHCS2 Yeast (Saccharomyces cerevisiae) was engineered to produce olivetolic acid by using CsHCS1 and CsHCS2 to synthesize hexanoyl-CoA, and a fusion of the cannabis olivetol synthase/polyketide synthase (PKS) and olivetolic acid synthase (O AS) to form olivetolic acid.
OAS was cloned in frame with the olivetol synthase/PKS using a synthetic linker sequence encoding the amino acids AATSGSTGSTGSTGSGRSTGSTGSTGSGRSHMV (SEQ ID NO:11) in the pESC-Trp yeast expression vector (Stratagene) under control of the GAL10 promoter. The open reading frame of CsHCS1 was cloned into the yeast expression vector pYESDEST52-Ura ( I nvitrogen) using Gateway technology. The open reading frame of CsHCS2 was cloned into pESC-Trp yeast expression vector (Stratagene) under control of the GAL1 promoter.
Yeast cells (InVSc I, Invitrogen) were transformed with the above constructs (OAS:olivetol synthase/PKS fusion alone, OAS:o livetol synthase/PKS fusion and CsHCS1; OAS:olivetol synthase/PKS fusion and CsHCS2) and the transformants grown on a SD-Trp plate for 3 days at 28°C. For each, a single colony was inoculated into 3 mL of SD-Trp glucose medium and incubated with shaking at 28°C for 2 days. A 0.5 mL aliquot of starter culture was used to inoculate 10 mL of SD-Trp galactose medium containing 1 mM sodium hexanoate and incubated at 20°C for 4 days. The complete culture was extracted with ethyl acetate, dried and the residue resuspended in 100 µL of 30% acetonitrile/70% water/0.05% formic acid. The products were analyzed using LCMS ( see Figure 5) .
Example 6:Role of CsHSC1 and CsHSC2 in Cannabinoid Biosynthesis in Plants Through a sequence-based analysis of the trichome EST dataset and biochemical assay of five AAEs, two that possess hexanoyl-CoA synthetase activity were identified (CsHSC1 and CsHSC2). To determine which of these is likely to be involved in the cannabinoid biosynthetic pathway, qRT-PCR and sub-cellular localization experiments were performed. qRT-PCR of CsHSC1 and CsHSC2 expression F inola plants were grown from seed until mid -flowering stage. Roots, stems, leaves, female flowers (w ith trichomes and after trichome isolation using the Beadbeater) , trichome cells and male flowers were sampled from three plants. Total RNA was isolated as described above. RNA had an Abs :Abs of >1.9 and showed distinct ribosomal 260 280 bands on denaturing gel. First-strand cDNA were synthesized using 0.5 µg RNA with a QuantiTect cDNA Synthesis kit ( Q iagen). Each 20 µL cDNA sample was diluted 1:4 with water, and 1 µL used as a PCR template. Gene-specific primers were designed to produce amplicons of 90-200 bp. PCR reactions (2 0 µL) were performed in 96-well plates using a SYBR Green based assay ( Q uantiFast SYBR Green kit, Qiagen) with a StepOne Plus instrument (Applied Biosystems). The cycling parameters used were 95°C for 5 min followed by 40 cycles of 95°C for 10 s, 60°C for 30 s, and a standard dissociation protocol (95°C 15 s, 60°C for 1 min, 60-95°C in 0.3°C increments for 15 s). Experiments were performed using cDNAs from three plants with two technical replicates. Actin, which was found to have stable expression in all tissues tested, was used as a reference gene. The efficiencies for all primer pairs were 90-110% as calculated using the standard curve method. C values were calculated using StepOne Software ( Applied Biosystems). The ­ Ct 2 method was used for relative gene expression analysis.
Subcellular localization of CsHSC1 and CsHSC2 YFP:CsHSC1 and YFP:CsHSC2 fusions were constructed by recombination into pEARLYGATE104 (Earley, K.W., Haag, J.R., Pontes, O., Opper, K., Juehne, T., Song, K. and Pikaard, C.S. (2006). Gateway-compatible vectors for plant functional genomics and proteomics. Plant J. 45, 616-629.) using LR recombinase (I nvitrogen). To generate an OLS:CFP construct, OLS lacking a stop codon was cloned into pCR8/GW/TOPO before recombination into pEARLYGATE102 using LR recombinase. The peroxisome marker PX-CK (Nelson, B.K., Cai, X. and Nebenführ, A. (2 007). A multicolored set of in vivo organelle markers for co-localization studies in Arabidopsis and other plants. Plant J. 51, 1126-1136.) was from ABRC (www.arabidopsis.org) . Plasmids were transformed into Agrobacterium tumefaciens GV3101 by electroporation and selected on LB plates containing 10 µg/mL rifampacin and 50 µg/mL kanamycin. Leaves of two-week old Nicotiana benthamiana plants were infiltrated with the Agrobacterium solution at an OD of 0.02 (Sparkes, I.A., Runions, J., Kearns, A. and Hawes, C. ( 2006). Rapid, transient expression of fluorescent fusion proteins in tobacco plants and generation of stably transformed plants. Nat. Protocol. 1, 2019-2025.). Two days post-infiltration, leaf epidermal cells were visualized using a Zeiss LSM510 confocal microscope. CFP was visualized with excitation 458 nm and image collection with a 475-525 nm bandpass filter; YFP at 514 nm with a 530-600 nm bandpass filter. Images were collected and analyzed using the Zeiss LSM software package.
The data provides evidence that CsHSC1 is the enzyme involved in cannabinoid biosynthesis. CsHSC1 was the most abundant transcript in the EST catalog, and qRT- PCR data shows that its expression is over 100-fold higher in trichome cells compared to other tissues (Figure 6). Furthermore, CsHSC1 is localized to the cytosol as evidenced by the sub-cellular localization experiment, which is the same compartment where the putative cannabinoid enzyme OLS is localized. The substrate preference of CsHSC1 provides additional evidence for its role in cannabinoid biosynthesis since it shows more specificity for hexanoate and other short-chain fatty acyl CoAs than CsHSC2 (Figure 3).
Although CsHSC1 is the enzyme likely involved in cannabinoid biosynthesis in plants, CsHSC2 is more efficient than CsHSC1 at synthesizing hexanoyl-CoA. However, CsHSC2 is localized to the peroxisome and it is not clear how hexanoyl-CoA formed in this compartment could be exported to the cytoplasm where the polyketide synthesis phase of the cannabinoid pathway is located. CsHSC2 accepts a very broad range of substrates, indicating that it is a more generalized acyl-CoA synthetase that may function in peroxisomal -oxidation.
Both CsHSC1 and CsHSC2 are valuable industrial tools. Knocking out CsHSC1 in cannabis plants could lead to a major reduction in cannabinoid levels in the plant, which is very desirable for hemp breeders. Over-expression of CsHSC1 in cannabis could lead to elevated cannabinoid levels, which is useful for pharmaceutical purposes. On the other hand, CsHSC2 would be particular useful in reconstituting cannabinoid formation in microorganisms or in an in vitro system.
Example 7:Generation of Mutants in the CsHSC1 and CsHSC2 genes using Targeted Induced Local Lesions IN Genomes (TILLING) Identification of cannabis plants with mutations in the CsHSC1 or CsHCS2 genes can be accomplished using TILLING. A mutagenized population of cannabis plants is screened using oligonucleotide primers and PCR in order to amplify the genes of interest.
Amplified mutant genes are annealed to wild-type genes to find mismatches between the mutant genes and the wild-type genes. Detected differences are used to identify plants that contain mutations in one of both of the CsHSC1 or CsHCS2 genes. Plants containing mutations that lead to altered amino acids in positions that are essential for the stability or alkanoyl-CoA synthetase activity of CsHSC1 or CsHCS2 proteins are unable to produce alkanoyl-CoA precursors for cannabinoid biosynthesis. The resulting plants contain reduced or altered levels cannabinoid products.
The present invention provides genes which encode two alkanoyl CoA synthetase enzymes from cannabis. These genes could be used to create, through breeding, targeted mutagenesis or genetic engineering, cannabis plants with enhanced cannabinoid production. In addition, inactivating or silencing a gene of the invention in cannabis could be used to block cannabinoid biosynthesis and thereby reduce production of cannabinoids such as THCA, the precursor of THC, in cannabis plants (e.g. industrial hemp). The genes of the present invention could be used, in combination with genes encoding other enzymes in the cannabinoid pathway, to engineer cannabinoid biosynthesis in other plants or in microorganisms or in cell-free systems, or to produce analogs of cannabinoid compounds or analogs of cannabinoid precursors.
Throughout the present disclosure, reference is made to publications, contents of the entirety of each of which are incorporated by this reference.
Other advantages that are inherent to the invention are obvious to one skilled in the art. The embodiments are described herein illustratively and are not meant to limit the scope of the invention as claimed. Variations of the foregoing embodiments will be evident to a person of ordinary skill and are intended by the inventors to be encompassed by the following claims.

Claims (29)

Claims:
1. An isolated or purified nucleic acid molecule comprising a nucleotide sequence having at least 75% sequence identity to SEQ ID NO:1 or a codon degenerate nucleotide sequence thereof, wherein the nucleic acid molecule encodes a polypeptide having alkanoyl-CoA activity.
2. The nucleic acid molecule of Claim 1, wherein the nucleotide sequence is as set forth in SEQ ID NO:1 or a codon degenerate nucleotide sequence thereof.
3. An isolated or purified nucleic acid molecule comprising a nucleotide sequence having at least 75% sequence identity to SEQ ID NO:3 or a codon degenerate nucleotide sequence thereof, wherein the nucleic acid molecule encodes a polypeptide having alkanoyl-CoA activity.
4. The nucleic acid molecule of Claim 3, wherein the nucleotide sequence is as set forth in SEQ ID NO:3 or a codon degenerate nucleotide sequence thereof.
5. The isolated or purified nucleic acid molecule of any one of Claims 1 to 4, wherein the nucleic acid molecule is cDNA.
6. The isolated or purified nucleic acid molecule of any one of Claims 1 to 5, further comprising a label, optionally a fluorescent label.
7. An isolated or purified polypeptide comprising an amino acid sequence having at least 85% sequence identity to SEQ ID NO:2 or a conservatively substituted amino acid sequence thereof, wherein the polypeptide has alkanoyl-CoA activity.
8. An isolated or purified polypeptide comprising an amino acid sequence having at least 85% sequence identity to SEQ ID NO:4 or a conservatively substituted amino acid sequence thereof, wherein the polypeptide has alkanoyl-CoA activity.
9. The polypeptide of Claim 7 or 8, further comprising a label, optionally a fluorescent label.
10. A vector, construct or expression system comprising the nucleic acid molecule of any one of Claims 1 to 6.
11. A host cell transformed with the nucleic acid molecule of any one of Claims 1 to 6, wherein the host cell is not within a human body.
12. A process of synthesizing an alkanoyl-CoA comprising: reacting carboxylic acid with CoA in presence of the polypeptide of any one of Claims 7 to 9.
13. The process of Claim 12, wherein the carboxylic acid has from 2 to 10 carbon atoms.
14. The process of Claim 12, wherein the alkanoyl-CoA comprises hexanoyl-CoA.
15. A process of altering levels of cannabinoid compounds in a cannabis plant, cannabis cell or cannabis tissue comprising using the nucleic acid molecule of any one of Claims 1 to 6, or a part thereof, to silence in the cannabis plant, cannabis cell or cannabis tissue a gene that encodes an enzyme that catalyzes synthesis of an alkanoyl-CoA, in comparison to a similar variety of organism, cell or tissue grown under similar conditions but without the use of the nucleic acid molecule for silencing.
16. A process of altering levels of cannabinoid compounds in a non-human organism, cell or tissue comprising mutating genes in the non-human organism, cell or tissue, and using the nucleic acid molecule of any one of Claims 1 to 6 to select for organisms, cells or tissues containing mutants or variants of a gene that encodes an enzyme that catalyzes synthesis of an alkanoyl-CoA.
17. A process of altering levels of cannabinoid compounds in a non-human organism, cell or tissue comprising expressing or over-expressing an exogenous nucleic acid molecule of any one of Claims 1 or 6 in the non-human organism, cell or tissue, in comparison to a similar variety of organism, cell or tissue grown under similar conditions but without the expressing or over-expressing of the exogenous nucleic acid molecule.
18. A process of altering levels of cannabinoid compounds in a non-human organism, cell or tissue comprising expressing or over-expressing an exogenous nucleic acid molecule encoding the polypeptide of any one of Claims 7 to 9 in the non-human organism, cell or tissue, in comparison to a similar variety of organism, cell or tissue grown under similar conditions but without the expressing or over-expressing of the exogenous nucleic acid molecule.
19. The process of Claim 17 or 18, wherein the level of the cannabinoid compound is increased.
20. The process of Claim 17 or 18, wherein the level of the cannabinoid compound is decreased.
21. A process of synthesizing a naturally-occurring cannabinoid compound or a non- naturally occurring analog of a cannabinoid compound in a non-human organism, cell or tissue comprising introducing and expressing the nucleic acid molecule of any one of Claims 1 to 6 in the non-human organism, cell or tissue in the presence of a carboxylic acid and CoA.
22. The process of any one of Claims 17 to 21, wherein the non-human organism is a microorganism.
23. The process of Claim 22, wherein the microorganism is Saccharomyces cerevisiae yeast or E. coli.
24. The process of any one of Claims 17 to 23, wherein the nucleic acid molecule is expressed or over-expressed in combination with expression or over-expression of one or more other nucleic acids that encode one or more enzymes in a cannabinoid biosynthetic pathway.
25. The process of Claim 24, wherein the one or more enzymes in a cannabinoid biosynthetic pathway is one or more of an olivetolic acid synthase, a type III polyketide synthase, a polyketide cyclase, an aromatic prenyltransferase or a cannabinoid-forming oxidocylase.
26. The process of Claim 25, wherein the one or more enzymes in a cannabinoid biosynthetic pathway is one or more of a type III polyketide synthase/olivetol synthase, a geranylpyrophosphate:olivetolate geranyltransferase, a -tetrahydrocannabinolic acid synthase, a cannabidiolic acid synthase or a cannabichromenic acid synthase.
27. The process of any one of Claims 16 to 26, wherein the cannabinoid compound is one or more of cannabigerolic acid, -tetrahydrocannabinolic acid, cannabidiolic acid, cannabichromenic acid, -tetrahydrocannabinol, cannabidiol or cannabichromene or a cannabinoid compound analog thereof comprising a side-chain of 1 to 9 carbon atoms in length.
28. A process of synthesizing an alkanoyl-CoA in an in vitro cell-free reaction, said process comprising: reacting a carboxylic acid with coenzyme A through the action of the polypeptide of any one of Claims 7 to 9.
29. An isolated or purified nucleic acid molecule according to any one of Claims 1 to 6 substantially as herein described with reference to the
NZ621037A 2011-07-13 2012-07-13 Genes and proteins for alkanoyl-coa synthesis NZ621037B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161507331P 2011-07-13 2011-07-13
US61/507,331 2011-07-13
PCT/CA2012/000656 WO2013006953A1 (en) 2011-07-13 2012-07-13 Genes and proteins for alkanoyl-coa synthesis

Publications (2)

Publication Number Publication Date
NZ621037A NZ621037A (en) 2016-02-26
NZ621037B2 true NZ621037B2 (en) 2016-05-27

Family

ID=

Similar Documents

Publication Publication Date Title
US9546362B2 (en) Genes and proteins for alkanoyl-CoA synthesis
US11306335B2 (en) Genes and proteins for aromatic polyketide synthesis
CA2770774C (en) Aromatic prenyltransferase from cannabis
EP2234474B1 (en) Diacylglycerol acyltransferase 2 genes and proteins encoded thereby from algae
Liu et al. Increasing seed mass and oil content in transgenic Arabidopsis by the overexpression of wri1-like gene from Brassica napus
NZ621037B2 (en) Genes and proteins for alkanoyl-coa synthesis
CA2724995A1 (en) Reduction of lyso-phosphatidylcholine acyltransferase activity
WO2010132988A1 (en) Increased seed oil and abiotic stress tolerance mediated by plant chd3 protein