NZ738299B2 - Production of noscapine - Google Patents

Production of noscapine

Info

Publication number
NZ738299B2
NZ738299B2 NZ738299A NZ73829916A NZ738299B2 NZ 738299 B2 NZ738299 B2 NZ 738299B2 NZ 738299 A NZ738299 A NZ 738299A NZ 73829916 A NZ73829916 A NZ 73829916A NZ 738299 B2 NZ738299 B2 NZ 738299B2
Authority
NZ
New Zealand
Prior art keywords
seq
plant
set forth
modified
nucleic acid
Prior art date
Application number
NZ738299A
Other versions
NZ738299A (en
Inventor
Ian Graham
Tracy Walker
Thilo Winzer
Original Assignee
Sun Pharmaceutical Industries Australia Limited
The University Of York
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
Priority claimed from GBGB1511156.0A external-priority patent/GB201511156D0/en
Application filed by Sun Pharmaceutical Industries Australia Limited, The University Of York filed Critical Sun Pharmaceutical Industries Australia Limited
Publication of NZ738299A publication Critical patent/NZ738299A/en
Publication of NZ738299B2 publication Critical patent/NZ738299B2/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
    • C12N15/63Introduction 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
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0071Oxidoreductases (1.) acting on paired donors with incorporation of molecular oxygen (1.14)

Abstract

This disclosure relates to high noscapine producing plants comprising functional or a non-functional or partially functional copy of a cytochrome P450-oxidoreductase catalysing the conversion of (S)-reticuline to (R)-reticuline in plants of the genus Papaver.

Description

WO 07643 Production of Noscapine Field of the Invention This disclosure relates to high noscapine producing plants comprisingfunctional, or a non-functional or partially functional copy of a cytochrome xidoreductase catalysing the conversion of (S)-reticuline to (H)-reticuline in plants of the genus Papaver.
Background to the Disclosure r somniferum, the opium poppy, is a source of clinically useful opiate alkaloids such as morphine, codeine, thebaine, ine and papaverine. Opiate alkaloids have broad clinical application ranging from analgesics to cough suppressants and anti- spasmodics and are known for inducing physical dependencies.
Opiate ids are structurally diverse with benzylisoquinoline providing the structural backbone and include nds such as papaverine, noscapine, codeine, apomorphine, bernerine and sanguinarine. isoquinoline alkaloids can be further grouped based on their chemical structure into morphinans and phthalideisoquinoline ids. Morphine, codeine, and thebaine are examples of morphinans, whereas papaverine and noscapine belong to the class of phthalideisoquinolines. Unlike morphinans noscapine has no analgesic properties but found its application as cough suppressant and acts as a potent antitumor agent binding to tubulin, inhibiting its polymerisation, arresting cell division and inducing apoptosis. The anti-tumor activity of noscapine and its derivatives was shown in various forms of cancer. Interestingly, contrary to most other opiate ids, ine does not induce physical dependencies.
Benzylisoquinoline alkaloids are found in a number of species in the Papaveraceae family, but morphinan production has only been reported in opium poppy and the closely related P. set/gerum. Total al synthesis, although possible, is not an economically viable means of production. Consequently, nan alkaloids are still exclusively sourced from the opium poppy plant. Whilst the biosynthesis of morphinans has been extensively studied over the last 25 years and the majority of genes in the pathway have been identified, the genes involved in the first step of morphinan synthesis, the epimerization of (S)-to (R)-reticuline, required for the development of efficient and economic viable morphinan production in microbial based s have just recently been ered in P. somniferum and are disclosed in patent application PCT/GBZO‘I5/051446 (W02015173590). ticuline is transformed to (R)-reticuline via 1,2—Dehydroreticuline by a fusion protein encoding both a cytochrome P450 and oxidoreductase module, the latter ing to the aldo-keto reductase family. Metabolite analysis of mutant alleles and heterologous expression demonstrate that the P450 module is sible for the conversion of (S)-reticuline to hydroreticuline while the oxidoreductase module converts 1,2—dehydroreticuline to (R)-reticuline rather than functioning as a P450 redox partner. A similar fusion protein catalyzing the conversion from (S)- to (R)-reticuline has also been described in Likewise, noscapine synthesis has only been reported in a number of Papaver species.
The level of noscapine in high noscapine varieties is typically less than 3 % of the capsule dry weight (which corresponds to a noscapine concentration of less than 60% of the total benzylisoquinoline alkaloid content) and vely low and when compared to the morphinan levels which constitute over 41 % of the capsule dry weight.
W02009/143574 and W02009/109012 ses P. somniferum with noscapine concentrations based on the alkaloid concentration in poppy straw of between 0.1 to a maximum of 3.1 %. The production of noscapine was found to require the presence of a complex cluster of co-expressed genes comprising mostly cytochrome P450 and methyltransferases converting scoulerine via several steps to noscapine and is disclosed in U82015/0004659, the content of which is incorporated by reference in its entirety.
This disclosure relates to the identification and characterization of high noscapine opium poppy plants sing a non-functional n of the P450-oxidoreductase fusion protein involved in the conversion of (S) to (R)—reticuline. Plants having the mutated gene have enhanced noscapine concentrations of over 3 % noscapine of the capsule dry weight (over at least 50% noscapine of the total benzylisoquinoline alkaloid content).
Poppy plants sing the modified fusion gene or engineered plants that are either null or have reduced expression of the wild-type fusion gene provide an alternative, more efficient source of noscapine.
Statements of Invention According to an aspect of the invention there is provided a plant of the genus r wherein said plant is modified in a gene ng a cytochrome P450-oxidoreductase catalysing the conversion of (S)-reticuline to (H)-reticuline wherein said modification is stably inherited and reduces or abrogates the expression or activity of said cytochrome P450 domain and/or oxidoreductase domain of said modified polypeptide compared to a wild-type plant comprising a functional cytochrome P450-oxidoreductase.
In an embodiment of the invention said plant is modified in a gene comprising a nucleotide sequence selected from the group ting of: i) a nucleic acid molecule sing a nucleotide sequence as set forth in SEQ ID NO: 1, 2 or 3; ii) a c acid molecule comprising a nucleotide sequence as set forth in SEQ ID NO: 4 or 33; iii) a nucleic acid molecule comprising a nucleotide sequence as set forth in SEQ ID NO: 5; iv) a nucleic acid molecule comprising a nucleic acid sequence as set forth in SEQ ID NO: 6, 7 or 8; v) a c acid molecule the complementary strand of which hybridizes under stringent hybridization ions to the nucleotide sequence set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33 and which encodes a polypeptide that has reduced or abrogated expression or activity of a cytochrome P450-oxidoreductase catalysing the conversion of (S)- reticuline to ticuline compared to a wild-type Papaver plant comprising a gene encoding a functional cytochrome P450- oxidoreductase ization of a nucleic acid molecule occurs when two complementary nucleic acid molecules undergo an amount of hydrogen bonding to each other. Isolated nucleic acid molecules as referred herein include genomic DNA, cDNA molecules and RNA les. The ency of hybridization can vary according to the environmental conditions surrounding the nucleic acids, the nature of the hybridization method, and the composition and length of the nucleic acid molecules used. Calculations regarding hybridization conditions required for attaining particular degrees of stringency are sed in Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2001); and Tijssen, Laboratory Techniques in Biochemistry and Molecular y—Hybridization with Nucleic Acid Probes Part I, Chapter 2 (Elsevier, New York, 1993). The Tm is the temperature at which 50% of a given strand of a nucleic acid molecule is hybridized to its complementary strand. The following is an exemplary set of hybridization conditions and is not limiting: Veg) High Stringency (allows sequences that share at least 90% identity to hybridize) Hybridization: 5x SSC at 65°C for 16 hours Wash twice: 2x SSC at room temperature (RT) for 15 minutes each Wash twice: 0.5x SSC at 65°C for 20 s each High Stringency gallows sequences that share at least 80% identity to hybridize) Hybridization: 5x-6x SSC at 65°C-70°C for 16-20 hours Wash twice: 2x SSC at RT for 5-20 s each Wash twice: 1x SSC at 55°C-70°C for 30 minutes each Low Stringency (allows sequences that share at least 50% identity to ize) ization: 6x SSC at RT to 55°C for 16-20 hours Wash at least twice: 2x-3x SSC at RT to 55°C for 20—30 minutes each.
In an embodiment of the invention said modified nucleic acid molecule is at least 50% identical to the nucleotide ce set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33.
In an embodiment of the ion the modified nucleic acid molecule is at least 55%, 60%, 65%, 70%, 75%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the nucleotide ce set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33 over the full nucleotide sequence.
In an embodiment of the invention said modified nucleic acid molecule is at least 99% identical to the nucleotide sequence set forth in SEQ ID NO 6, 7 or 8, over the full nucleotide sequence and wherein said modification is an addition, substitution or deletion of at least one tide base between nucleotides 1280-1300 set forth in SEQ ID NO 6, 7 or 8.
In an embodiment of the invention said modification is a substitution of at least one nucleotide base between nucleotides 1280-1300 set forth in SEQ ID NO 6, 7 or 8.
In an embodiment of the invention said modification is a substitution of at least one nucleotide base between nucleotides 1285-1295 set forth in SEQ ID NO 6, 7 or 8.
In an embodiment of the invention said modification is a C to T substitution of the nucleotide base at nucleotide 1291 of SEQ ID NO 6, 7 or 8.
In a preferred embodiment of the invention there is provided a modified polypeptide encoded by c acid molecule comprising a nucleotide sequence as herein disclosed.
In an embodiment of the invention said plant expresses a modified P450-oxidoreductase polypeptide wherein said modified polypeptide has reduced or ted activity in the conversion of (S)-reticuline to (H)-reticuline.
In an embodiment of the invention said modified polypeptide is selected from the group consisting of: i) a polypeptide comprising a modified amino acid sequence of the amino acid sequence set forth in SEQ ID NO: 9, 10 or 11; a polypeptide comprising a modified amino acid sequence of the amino acid ce as set forth in SEQ ID NO: 12, 13 or 14; a polypeptide comprising a modified amino acid sequence that is at least 95% identical to the full length amino acid sequence as set forth in SEQ ID NO: 9, 10, 11, 12, 13 or 14, wherein said sequence is modified by addition, deletion or substitution of one or more amino acid sequences and wherein said polypeptide has reduced or abrogated enzyme activity in the conversion of ticuline to 1,2-dehydroreticuline and/or to (H)- line when compared to the unmodified polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 9, 10, 11, 12, 13 or 14.
In an embodiment of the invention said modified polypeptide is ed from the group ting of: i) a modified polypeptide comprising a modified amino acid sequence of the amino acid sequence set forth in SEQ ID NO: 15; a modified polypeptide comprising a modified amino acid ce of the amino acid sequence set forth in SEQ ID NO: 16; a modified polypeptide sing a modified amino acid sequence that is at least 95% cal to the full length amino acid sequence as set forth in SEQ ID NO: 15 or 16, wherein said sequence is modified by addition, deletion or substitution of one or more amino acid sequences and wherein said polypeptide has reduced or abrogated enzyme activity in the conversion of 1,2-dehydroreticuline to (H)-reticuline when compared to the unmodified polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 15 or 16.
In an embodiment of the ion said ed polypeptide comprises a deletion or substitution of amino acid e 431 of SEQ ID NO: 9 wherein said modified polypeptide is at least 90% identical to the amino acid sequence set forth in SEQ ID NO: In an ment of the invention said modified polypeptide comprises a on or substitution of amino acid residue 432 of SEQ ID NO: 10 wherein said modified polypeptide is at least 90% identical to the amino acid sequence set forth in SEQ ID NO: 10.
In an embodiment of the invention said modified ptide comprises a deletion or substitution of amino acid residue 431 of SEQ ID NO: 11 wherein said modified polypeptide is at least 90% identical to the amino acid sequence set forth in SEQ ID NO:11.
In an embodiment of the invention said modified polypeptide is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid ce set forth in SEQ ID NO: 9 n said modified polypeptide comprises a deletion or substitution of amino acid residue 431 of SEQ ID NO: 9.
In an embodiment of the invention said modified polypeptide is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence set forth in SEQ ID NO: 10 wherein said modified polypeptide comprises a deletion or substitution of amino acid residue 432 of SEQ ID NO: 10.
In an embodiment of the invention said modified polypeptide is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence set forth in SEQ ID NO: 11 wherein said modified polypeptide comprises a deletion or substitution of amino acid residue 431 of SEQ ID NO: 11.
In an embodiment of the invention said modified polypeptide comprises a substitution or on at amino acid arginine 431 of the amino acid sequence set forth in SEQ ID NO: In an ment of the invention said modified polypeptide comprises a substitution or deletion at amino acid arginine 432 of the amino acid sequence set forth in SEQ ID NO: In an embodiment of the invention said modified polypeptide comprises a substitution or deletion at amino acid arginine 431 of the amino acid sequence set forth in SEQ ID NO: In an embodiment of the ion said modification is the substitution of arginine for tryptophan in at position 431 of the amino acid sequences set forth in SEQ ID NO: 9.
In an embodiment of the invention said modification is the substitution of arginine for tryptophan in at position 432 of the amino acid sequences set forth in SEQ ID NO: 10.
In an embodiment of the invention said modification is the substitution of ne for tryptophan in at position 431 of the amino acid sequences set forth in SEQ ID NO: 11.
In an embodiment of the invention said modified polypeptide comprises the amino acid sequence set forth in SEQ ID NO: 27.
In an embodiment of the invention said modified ptide comprises the amino acid sequence set forth in SEQ ID NO: 28.
In an embodiment of the invention said modified polypeptide comprises the amino acid sequence set forth in SEQ ID NO: 29.
In an embodiment of the invention said ed polypeptide is encoded by a nucleic acid molecule comprising a nucleotide sequence set forth in SEQ ID NO: 30.
In an ment of the invention said modified ptide is encoded by a nucleic acid molecule comprising a tide sequence set forth in SEQ ID NO: 31.
In an embodiment of the invention said modified polypeptide is encoded by a nucleic acid molecule comprising a nucleotide sequence set forth in SEQ ID NO: 32.
In a further embodiment of the ion said plant is heterozygous for said modified gene.
In an alternative ment of the invention said plant is gous for said modified gene.
Mutagenesis as a means to induce phenotypic changes in organisms is well known in the art and includes but is not limited to the use of mutagenic agents such as chemical mutagens (e.g. base analogues, deaminating agents, DNA intercalating agents, alkylating agents, transposons, bromine, sodium azide] and physical mutagens [e.g. ionizing radiation, psoralen exposure combined with UV ation). It is also common general knowledge that ablation of gene function can be obtained through genetic means such as siRNA and antisense. The introduction of double ed RNA, also referred to as small inhibitory/interfering RNA (siRNA) or short n RNA [shRNA], into a cell which s in the destruction of mRNA mentary to the sequence included in the siRNA/shRNA molecule. This is typically via transfection of expression ucts, for example those disclosed in , the content of which is incorporated in its entirety, which are stably integrated into the genome and inherited in a Mendelian In an embodiment of the invention said modified Papaver plant further comprises one or more genes comprising a nucleotide sequence selected from the group consisting of: i) a nucleotide sequence as represented by the sequence in SEQ ID NO:, 17,18, 19, 20, 21, 22, 23, 24, 25 or 26; ii) a nucleotide sequence wherein said sequence is degenerate as a result of the genetic code to the nucleotide sequence defined in (i); iii) a nucleic acid molecule the complementary strand of which hybridizes under stringent hybridization conditions to the sequence in SEQ ID NO: 17,18, 19, 20, 21, 22, 23, 24, 25 or 26 wherein said nucleic acid molecule encodes polypeptides ed in the biosynthesis of Papaver noscapine or intermediates in the biosynthesis of noscapine.
In an embodiment of the invention said nucleic acid molecule is at least 55%, 60%, 65%, 70%, 75%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the nucleotide sequence set forth in SEQ ID NO: 17,18, 19, 20, 21, 22, 23, 24, 25 or 26 over the full nucleotide sequence.
In an embodiment of the invention said plant comprises enhanced levels of noscapine or intermediates in the synthesis of noscapine.
In an embodiment of the invention noscapine comprises at least 50% of the benzylisoquinoline alkaloid content of said ed plant.
In a r ment of the invention said plant ses a noscapine content that is at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% of the benzylisoquinoline alkaloid content of said modified plant.
WO 07643 In an alternative embodiment of the invention said plant comprises a ine content that is at least 50% of the alkaloid content of a combination of alkaloids comprising morphine, codeine, oriparvine, thebaine and noscapine.
In a further embodiment of the invention said plant comprises a noscapine content that is at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% of the alkaloid t of a combination of alkaloids comprising morphine, codeine, vine, thebaine and noscapine.
In an ment of the invention said plant comprises a benzylisoquinoline content that comprises substantially noscapine.
In an embodiment of the invention said Papaver plant is Papaver somniferum.
According to an aspect of the invention there is provided a Papaver plant modified by transformation or transfection with an expression vector adapted to express an antisense, siRNA or shRNA designed with reference to a tide sequence set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33 n noscapine comprises at least 50% of the benzylisoquinoline alkaloid content of said modified plant.
In an embodiment of the ion said modified plant comprises a noscapine content that is at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% of the of the benzylisoquinoline alkaloid content of said modified plant.
In an embodiment of the invention said modified plant comprises a benzylisoquinoline alkaloid content that comprises substantially noscapine.
In an alternative embodiment of the invention said modified plant comprises a noscapine content that is at least 50% of the alkaloid content of a combination of alkaloids comprising morphine, codeine, oriparvine, thebaine and noscapine.
In a further embodiment of the invention said modified plant comprises a ine content that is at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% of the alkaloid t of a combination of alkaloids comprising morphine, codeine, vine, thebaine and noscapine.
According to a further aspect of the invention there is provided a Papaver capsule ed from the modified plant according to the invention.
According to a further aspect of the invention there is provided a Papaver straw obtained from the modified plant according to the invention.
According to an aspect of the invention there is provided a Papaver seed obtained from the ed plant according to the invention.
According to a further aspect of the invention there is provided a modified polypeptide comprising an amino acid sequence wherein said amino acid sequence is at least 90% identical to the amino acid ce set forth in SEQ ID NO: 27, 28 or 29.
According to a further aspect of the invention there is provided a ed polypeptide comprising an amino acid sequence wherein said amino acid sequence is at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence set forth in SEQ ID NO: 27, 28 or 29.
According to an aspect of the invention there is provided a ed ptide comprising the amino acid ce set forth in SEQ ID NO: 27.
According to an aspect of the invention there is provided a modified polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 28.
According to an aspect of the invention there is provided a modified polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 29.
According to a further aspect of the invention there is provided a c acid molecule sing a nucleotide sequence wherein said nucleotide sequence is at least 90% identical to the nucleotide sequence set forth in SEQ ID NO: 30, 31 or 32.
According to an aspect of the invention there is ed a nucleic acid molecule comprising a nucleotide sequence wherein said nucleotide sequence is at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the tide sequence set forth in SEQ ID NO: 30, 31 or 32.
According to an aspect of the invention there is provided a nucleic acid molecule comprising a nucleotide sequence set forth in SEQ ID NO: 30.
According to an aspect of the invention there is provided a nucleic acid molecule comprising a nucleotide sequence set forth in SEQ ID NO: 31.
According to an aspect of the invention there is provided a nucleic acid molecule sing a nucleotide sequence set forth in SEQ ID NO: 32. ing to a further aspect of the invention there is provided a method to obtain a Papaver plant comprising enhanced or elevated noscapine, or intermediates in the synthesis of noscapine, comprising the steps of: i) mutagenesis of wild-type seed from a Papaver plant that comprises a gene encoding a functional cytochrome P450-oxidoreductase catalysing the conversion of (S)-reticuline to (H)—reticuline; ii) cultivation of the seed in i) to e first and uent tions of plants; iii) obtaining seed from the first tion plant and subsequent generations of plants; iv) determining if the seed from said first and uent generations of plants has altered nucleotide sequence of said gene encoding a cytochrome P450—oxidoreductase; v) obtaining a sample and analysing the nucleic acid sequence of a nucleic acid molecule selected from the group consisting of: a) a nucleic acid le comprising a nucleotide sequence as represented in SEQ ID NO: 1, 2, 3,4, 5,6, 7, 8 or 33; b) a nucleic acid le that hybridises to a complementary strand of said nucleic acid molecule in a) under stringent hybridisation conditions and that encodes a non-functional cytochrome P450-oxidoreductase; and optionally vi) comparing the nucleotide sequence of the nucleic acid molecule in said sample to a nucleotide ce of a nucleic acid molecule of the original wild-type plant.
In an embodiment of the method of the invention said plants have an altered expression of said rome P450-oxidoreductase.
In a further embodiment of the method of the invention said modified nucleic acid molecule is at least 55%, 60%, 65%, 70%, 75%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the nucleotide sequence set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33 over the full nucleotide sequence.
In a yet further embodiment of the method of the invention said nucleic acid molecule is analysed by a method comprising the steps of: i) ting nucleic acid from said mutated plants; ii) amplification of a part of said c acid molecule by a polymerase chain reaction; iii) forming a preparation comprising the ied c acid and nucleic acid extracted from wild-type seed to form duplex nucleic acid; iv) incubating said preparation with a single stranded nuclease that cuts at a region of heteroduplex nucleic acid to identify the mismatch in said heteroduplex; and v) determining the site of the mismatch in said nucleic acid heteroduplex.
In an ment of the method of the invention said modified plant is ed for noscapine content and optionally compared to the original unmodified plant.
In an embodiment of the method of the invention said Papaver plant is Papaver somniferum. ing to a further aspect of the invention there is provided a plant obtained by the method according to the invention.
According to a further aspect of the invention there is provided the use of a gene encoding a cytochrome P450-oxidoreductase catalysing the conversion of (S)-reticuline to (H)-reticuline in the fication of Papaver cultivars that have enhanced levels of noscapine or intermediates in noscapine synthesis.
In an embodiment of the invention said gene comprises a nucleotide sequence selected from the group consisting of: i) a nucleic acid le comprising a nucleotide sequence as set forth in SEQ ID NO: 1, 2 or 3; ii) a nucleic acid molecule comprising a nucleotide sequence as set forth in SEQ ID NO: 4 or 33; iii) a nucleic acid molecule comprising a nucleotide sequence as set forth in SEQ ID NO: 5; iv) a nucleic acid molecule comprising a nucleic acid sequence as set forth in SEQ ID NO: 6, 7 or 8; a nucleic acid molecule the complementary strand of which hybridizes under stringent hybridization conditions to the nucleotide ce set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 33 and which encodes a polypeptide that has reduced or abrogated expression or activity of a cytochrome P450-oxidoreductase sing the conversion of (S)- reticuline to (H)-reticuline compared to a wild-type Papaver plant comprising a gene encoding a functional cytochrome P450- oxidoreductase.
According to a further aspect of the invention there is provided a method to identify a Papaver plant comprising a modified cytochrome P450-oxidoreductase gene comprising the steps: obtaining a sample of plant material from a plant to be tested; extracting genomic nucleic acid from said plant material; analysing the extracted genomic nucleic acid encoding a variant nucleotide sequence of the nucleotide sequences set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33; and optionally, comparing the ce to a wild-type nucleotide sequence comprising SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33.
Competitive allele specific PCR (KASP) allows the identification of single-nucleotide polymorphism in a DNA sequence. The method is based on primer extension of two allele-specific (fonNard) s and amplification of the resulting product using a common reverse primer. The forward primers are designed specifically to bind and extend the primer to either the SNP or wild type allele which allows subsequent binding of the common reverse primer for amplification of the t. In a second step an oligonucleotide cassette complimentary to a specific 5’ sequence of the first allele- specific d primer and a second oligonucleotide te complimentary to a specific 5’ sequence of the second allele-specific fonNard primer both comprising a different fluorescent label is added to the reaction allowing ication of the SNP and wild type targets and product detection through fluorescent signalling. Alternatively, ly scent labelled primers targeted to the common reverse primer can be used to amplify e.g. just the species with the SNP in a sample. The KASP technology is sed in US2016/0002712 which is incorporated by reference in its entirety.
In a red method of the invention said analysis comprises the steps: i) preparing a reaction mixture comprising: a) genomic DNA to be analysed, b) a first allele specific fonNard o|igonuc|eotide primer comprising a nucleotide ce complementary to said cytochrome P450-oxidoreductase gene nucleotide sequence and a nucleotide sequence complementary to a tide sequence of a first fluorescent reporter cassette, c) a second allele specific fonNard o|igonuc|eotide primer comprising a nucleotide sequence complementary to said cytochrome P450-oxidoreductase gene an a nucleotide sequence complementary to a nucleotide sequence of a second fluorescent reporter cassette wherein the nucleotide at the 3’-end of said second uc|eotide primer sequence is complimentary to a polymorphic nucleotide sequence variant of said cytochrome P450-oxidoreductase gene, and wherein the nucleotide sequence of said first and second uc|eotide s are complementary to the same upstream nucleotide sequence of said cytochrome P450-oxidoreductase gene; d) a reverse o|igonuc|eotide primer sing a nucleotide sequence complementary to a nucleotide sequence downstream of said cytochrome P450- oxidoreductase gene nucleotide sequence; and e) two reporter cassettes comprising double stranded nucleic acid sing: a first double stranded c acid wherein one strand comprises a fluorescent label and is complementary to said first allele specific primer and a second strand comprising a quencher label that quenches said fluorescent label; and a second double stranded nucleic acid wherein one strand comprises a fluorescent label different from the fluorescent label of i) and is complementary to said second allele specific primer and a second strand comprising a quencher label that quenches said second fluorescent label; ii) heating said reaction mixture to denature genomic c acid and said double stranded reporters and providing conditions to anneal s to their respective complementary nucleotide sequence, primer extending said annealed o|igonuc|eotide primers and polymerase chain amplification of said e; and iii) detecting fluorescence emission.
In a red method of the invention said modified and wild type cytochrome P450- oxidoreducatse gene nucleic acid sequence is as set forth SEQ ID NO: 6.
In a preferred method according of the invention said first fonNard primer comprises (SEQID NO: 35) CCAGGCAATCATCAAAGAATCAATGT (SEQ ID NO: 35), said second fonNard primer comprises CCAGGCAATCATCAAAGAATCAATGC (SEQ ID NO: 34) and said reverse primer comprises CACGGGGCTGGCTGGATACAA (SEQ ID NO In an embodiment of the invention said use or method es analysis of the sequence or expression of a further gene ed from the group consisting of: i) a nucleotide sequence as represented by the sequence in SEQ ID NO: 17, 18, 19 25 or 26; , 20, 21, 22, 23, 24, ii) a nucleotide sequence wherein said sequence is degenerate as a result of the genetic code to the nucleotide sequence defined in (i); iii) a nucleic acid molecule the complementary strand of which hybridizes under stringent ization conditions to the sequence in SEQ ID NO: 17, 18, 19 20, 21 or, 22, 23, 24, 25 or 26 wherein said nucleic acid le encodes polypeptides involved in the biosynthesis of Papaver noscapine or intermediates in the biosynthesis of noscapine.
In an embodiment of the invention said nucleic acid molecule is at least 55%, 60%, 65%, 70%, 75%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the nucleotide sequence set forth in SEQ ID NO: 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 over the full tide sequence.
It will be apparent to the skilled artisan that techniques that enable the analysis of mutations in gene ces and/or expression are available and include, but are not limited to genomic DNA sequencing, PCR based analysis of DNA via single polynucleotide polynucleotide polymorphisms (SNP analysis), single stranded conformational polymorphism (SSCP), ring nt gel electrophoresis (DGGE), quantitative PCR and multiplex PCR to detect expression of multiple genes.
According to a further aspect of the invention there is provided a method for the extraction of noscapine from a modified plant or capsule or straw according to the invention comprising the steps: i) harvesting a modified plant, capsule or straw ing to the invention and forming a reaction mixture of particulate plant al; ii) solvent extracting said reaction mixture to provide an alkaloid enriched fraction; and iii) concentrating said enriched alkaloid fraction to provide an enriched noscapine fraction.
Throughout the description and claims of this specification, the words "comprise" and "contain" and variations of the words, for example "comprising" and "comprises", means "including but not limited to", and is not intended to (and does not) exclude other moieties, additives, components, integers or steps. "Consisting essentially" means having the essential integers but including integers which do not materially affect the function of the essential integers.
Throughout the description and claims of this specification, the singular encompasses the plural unless the context othenNise es. In particular, where the indefinite article is used, the specification is to be understood as contemplating plurality as well as arity, unless the context es otherwise.
Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example bed herein unless incompatible therewith.
Definitions "Benzylisoquinoline alkaloids" typically include papaverine, noscapine, codeine, morphine, apomorphine, berberine, protopine, tubocurarine and narine. The major ids are morphine, codeine, oriparvine, thebaine and noscapine.
"Substantially noscapine" means that other isoquinoline alkaloids are trace alkaloids or undetectable isoquinoline alkaloids as measured by techniques available to the d person.
"Stably ted" refers to the transfer of nt characteristics which remain unchanged after repeated propagation or, in the case of a particular cycle of propagation, at the end of each such cycle.
"Reduced or abrogated expression" refers to reduction of mRNA expression by 10, 20, , 40, 50, 60, 70, 80, 90 or 100% when ed to a wild-type plant expressing a gene encoding an unmodified cytochrome p450-oxidoreductase. Quantitative mRNA expression methods are known to the skilled person such for example real time PCR. ional cytochrome P450-oxidoreductase" refers to a non-modified enzyme which is capable of ting (S)- to (R)- reticuline via 1,2—dehydroreticuline. The measurement of cytochrome P450 and eductase activities are well known in the art, for example see Lenz & Zenk, 1995, FEBS 233:132—139. In addition heterologous expression of cytochrome P450 and eductase domains in addition to expression of cytochrome xidoreductase fusion protein in microbial systems such as yeast allows functional characterisation of functional and non-functional cytochrome P450-oxidoreductase enzymes.
The phrase "enhanced levels of noscapine and noscapine intermediates" means producing a higher level of ine or intermediates in the synthesis of noscapine ed to a wild-type plant comprising a functional cytochrome P450-oxidoreductase, for example, a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98% or 99% increase in the level of noscapine or intermediates in noscapine biosynthesis.
"VWd-type plant" means a plant having a ype that is characteristic of the species in a natural breeding population. ed or abrogated activity in the sion of (S)-reticuline to (H)-reticuline" refers to the reduction of the catalytic activity of the enzyme by 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100% relative to an unmodified cytochrome P450-oxidoreductase.
Variant as referred to under "SEQ ID Summary" refers to different polymorphic forms of the ular gene.
SEQ ID NO: 1 P450 domain nucleotide sequence; polymorphic variant a SEQ ID NO: 2 P450 domain nucleotide sequence; polymorphic variant b SEQ ID NO: 3 P450 domain nucleotide sequence; polymorphic variant c SEQ ID NO: 4 oxidoreductase A nucleotide sequence, variant b SEQ ID NO: 5 oxidoreductase B nucleotide sequence SEQ ID NO: 6 cytochrome P450-oxidoreductase tide sequence; polymorphic variant a SEQ ID NO: 7 cytochrome P450-oxidoreductase nucleotide sequence; polymorphic variant b SEQ ID NO: 8 cytochrome P450-oxidoreductase nucleotide sequence; polymorphic variant c SEQ ID NO: 9 cytochrome P450-oxidoreductase amino acid, rphic variant a SEQ ID NO: 10 cytochrome P450-oxidoreductase amino acid; polymorphic variant b SEQ ID NO: 11 cytochrome P450-oxidoreductase amino acid; polymorphic variant c SEQ ID NO: 12 P450 domain amino acid; polymorphic variant a SEQ ID NO: 13 P450 domain amino acid; polymorphic variant b SEQ ID NO: 14 P450 domain amino acid; rphic variant c SEQ ID NO: 15 oxidoreductase A amino acid SEQ ID NO: 16 oxidoreductase B amino acid SEQ ID NO: 17 PSMT1 SEQ ID NO: 18 PSMT2 SEQ ID NO: 19 PSMT3 SEQ ID NO: 20 CYP450 1 (CYP 82X1) SEQ ID NO: 21 CYP450 2 (CYP 719A21) SEQ ID NO: 22 CYP450 3 (CYP 82X2) SEQ ID NO: 23 CYP450 4 (CYP 82Y1) SEQ ID NO: 24 PSAT1 SEQ ID NO: 25 PSSDR1 SEQ ID NO: 26 PSCXE1 SEQ ID NO: 27 cytochrome xidoreductase mutant amino acid, variant a SEQ ID NO: 28 cytochrome P450-oxidoreductase mutant amino acid, variant b SEQ ID NO: 29 cytochrome xidoreductase mutant amino acid, variant c SEQ ID NO: 30 cytochrome P450-oxidoreductase mutant nucleotide sequence variant a SEQ ID NO: 31 cytochrome P450-oxidoreductase mutant nucleotide sequence variant b SEQ ID NO: 32 cytochrome P450-oxidoreductase mutant nucleotide sequence variant c SEQ ID NO: 33 Oxidoreductase A, nucleic acid sequence, variant a (and c) An embodiment of the invention will now be described by example only and with reference to the figures, examples and tables: Figure 1: Violin plot of the relative amount (% total) of noscapine and morphinan alkaloids in dried capsules of F2 populations segregating storr—4. HOM_STORR4, F2 material homozygous for storr—4; ORR4, F2 al heterozygous for storr—4; VVT_STORR, F2 material homozygous for the STORR wild type allele; HN4, l plants of NOSCAPINE CVS 4, one of the parental lines of the F2 material; Figure 2: Violin plot of the absolute amount (% capsule dry weight) of noscapine and morphinan alkaloids in dried capsules of segregating F2 tions segregating storr—4.
HOM_STORR4, F2 material homozygous for storr—4; HET_STORR4, F2 al heterozygous for storr—4; VVT_STORR, F2 material homozygous for the STORR wild type allele; HN4, control plants of NOSCAPINE CVS 4, one of the parental lines of the F2 material; DW, dry weight; and Figure 3: Schematic overview of noscapine and morphinan biosynthetic pathways in opium poppy and the position of the pathway block conferred by storr—4. (S)-reticuline is a major branch point intermediate in alkaloid metabolism in opium poppy g as a precursor for both noscapine and morphinan biosynthesis. The STORR locus ls the gateway reaction into morphinan biosynthesis, the two step epimerisation of (S)- to ticuline. Impairment of STORR, for example by means of knock out mutations such as storr—4, leads to a block of morphinan biosynthesis and sed production of noscapine in noscapine producing cultivars of opium poppy.
Materials & s EMS mutagenesis Prior to mutagenesis, M0 seeds from P. somniferum cultivar GSK INE CV82 were soaked in 0.1% (w/v) potassium chloride solution for approximately 24 hours. The seed was then imbibed for three hours with ethyl methanesulfonate (EMS) solution (200 mM EMS in 100 mM sodium hydrogen phosphate buffer, pH 5.0, supplemented with 700 mM dimethyl sulfoxide). The treated M1seed was washed twice for 15 minutes each with 100 mM sodium thiosulphate solution and then twice for 15 s each with distilled water. A volume of 10 mL of the respective solutions was used per 1000 seeds and all steps were carried out on a rocking platform. The washed M1 seed was dried overnight on Whatman 3MM Blotting paper (Whatman/GE Healthcare Life Sciences, Little Chalfont, UK) and sown the following morning on top of a mixture of John lnnes no 2 compost, vermiculite and perlite (4:2:1), then covered with a thin layer of compost.
Seedlings were transplanted three to four weeks after germination into larger pots and grown in the glasshouse until maturity. Dried es were harvested by hand from the M1 population once capsules had dried to approximately 10% moisture on the plant. M2 seed was manually separated from the e, providing the M2 seed families that were used in the based trials described below.
Field-based trials of EMS-mutagenised lines M2 seed was sown in in the field in Tasmania in the 010 growing season. M2 seed lines from the EMS mutagenised population were hand sown in 3m long rows. After germination, plants were thinned to 75 mm apart. Typically 4 plants (designated A, B, C, D) per M2 line were self-pollinated (two re buds per plant selected) by securing a linen bag over the bud. ollinated rows were ted at ~ 11% moisture, and assayed (as described below) for morphine (M), codeine (C), oripavine (O) and thebaine (T) content. Analysis of a selected self-pollinated capsules from an M2 plants indicated significantly reduced morphinan assays, and significantly higher assays of noscapine.
M3 and M4 seeds derived from a selected self-pollinated M2 capsule displaying the low morphinan alkaloids/high noscapine phenotype were sown in the following growing seasons in Tasmania to confirm this phenotype in both open-pollinated and self- pollinated capsules. Typically 4 plants (designated A, B, C, D) were self-pollinated per M3 and M4 seed line as described above. GSK commercial ars were grown ide the M2, M3 and M4 lines as a comparison. Depending on the , they included GSK MORPHINE CULTIVARs 3 and 5, GSK THEBAINE CULTIVAR 3, GSK NOSCAPINE CULTIVARs 3 - 6.
Poppy straw is of based trials Poppy capsules were harvested from the respective mutant lines by hand once capsules had dried to approximately 10% moisture on the plant. The seed was manually separated from the capsule, and capsule straw material (poppy straw) was then shipped to the GSK extraction facility in Port Fairy, Australia.
The poppy straw samples were ground in a Retsch Model MM400 ball mill into a fine powder. Two gram samples of ground poppy straw (2 :r 0.003 g) were extracted in 50 mL of a 10% acetic acid solution. The extraction suspension was shaken on an l shaker at 200rpm for a minimum of 10 minutes then filtered to provide a clear te. The final filtrate was passed through a 0.22 um filter prior to analysis.
The solutions were analysed using a Waters Acquity UPLC system (Waters Corporation, Milford, MA, USA) fitted with a Waters Acquity BEH C18 column, 2.1mm x 100mm with 1.7 micron packing. The mobile phase used a gradient profile with eluent A ting of 0.1% Trifluoroacetic acid in deionised water and eluent B consisting of 100% Acetonitrile.
The mobile phase gradient conditions used are as listed in Table 1, the gradient curve number as determined using a Waters Empower tography software package. The flow rate was 0.6 mL per minute and the column ined at 45 degrees centigrade.
The injection volume was 1 uL injection volume and the ids were detected using a UV detector at 285 nm.
The loss on drying (LOD) of the straw was determined by drying in an oven at 105 degrees rade for 16-20 hours.
Table 1 — Gradient Flow Program % Eluent Flow TIME (minutes)_ % Eluent A Curve No B (lemin) __-IE- 3 __-IE- 6 __-IE- 11 Alkaloid concentrations for morphine, pseudomorphine, codeine, thebaine, oripavine and noscapine were quantified using external standard calibration and the results calculated on a dry weight basis. Reticuline was quantified using the thebaine calibration (assuming the ne response) and s calculated on a dry weight basis (% weight relative to thebaine; %WRT).
Typical retention times are as follows: Compound Retention Time (minutes) Morphine 1.14 Pseudomorphine 1.26 Codeine 1.69 Reticuline 2.05 -Hydroxythebaine 2.32 Thebaine 2.53 Noscapine 3.16 Leaf latex is of glasshouse grown M4 generation (S-186668) derived from the self-pollinated MCS 746 C plant.
The latex alkaloid profile ined in M4 siblings of the plants used for sequencing the cytochrome P450-oxidoreductase (STORH) cDNA (see below). Latex was collected when the first flower buds d (~7 week old plants) from cut petioles, with a single drop dispersed into 500 uL of 10% acetic acid. This was diluted 10x in 1% acetic acid to give an alkaloid solution in 2% acetic acid for further analysis. The samples were analysed by UPLC-MS and R-scripts as previously described ("the standard method"; (V\finzer T. et al. (2012) Science 336, )). Compounds were annotated by generation of empirical formulae from exact masses (< 5 ppm mass accuracy) and comparison to authentic standards. Unknowns were ted by a masstag in the format MxTy, where x is mass and y is retention time in seconds.
The relative content of alkaloids was measured as % peak area relative to total alkaloid peak area.
Isolation and cing the of the rome P450-oxidoreductase cDNA in the M4 generation 668) derived from the self-pollinated MCS 746 C plant.
RNA was isolated from M4 duals derived from self-pollinated M3 individual MCS 746 C that displayed the low morphinan alkaloid/high noscapine phenotype. M4 plants were grown under glasshouse conditions to post-flowering stages (1-6 days after petal fall) and stem segments (2.5-3 cm long) immediately beneath the flowers were harvested and flash frozen in liquid nitrogen.
Total RNA was extracted from the stems using the pine tree RNA extraction method (Chang et al., 1993. Plant Mol. Biol. Rep. 11: 113-116) and further purified using the RNeasy Plus MicroKit (Qiagen, Crawley, UK). sis of cDNA was performed using 1 pg of total RNA, 10 uM oligo dT MW 4500 (lnvitrogen/Life technologies, Paisley, UK) and 1 mM dNTP. Reactions were incubated for 5 min at 65°C to allow annealing of oligonucleotides. First strand synthesis reactions contained 1 X First Strand buffer (lnvitrogen/Life Technologies), 20 mM DTT, 40 U RNAse out (lnvitrogen/Life logies). Reactions were ted at 42°C for 2 min and then 200 U SuperScript || reverse transcriptase (lnvitrogen/Life technologies, Paisley, UK) was added followed by a 50 min incubation at 42°C and heat inactivation at 70°C for 15 min. Samples were diluted 5 X in water and used for gene specific amplifications.
The full-length region ng the cytochrome P450-oxidoreductase gene was amplified from the cDNA samples using the primers 5’- ATGGAGCTCCAATATATTTC-3’, 5’- TCAAGCTTCATCATCCCACA-3’ and the high fidelity Q5® Hot Start High-Fidelity DNA Polymerase (New England Biolabs, Hitchin, UK) according to the guidelines supplied by the manufacturer. Cycling conditions were as follows: 98°C for 30 s, 30 cycles of 98°C for 10 s, 60°C for 30 s, 72°C for 1.5 min and a final extension at 72°C for 2 min. The 3 kb-PCR product was then ligated into the pJET1.2 cloning vector using the CloneJET PCR Cloning Kit (Thermo Scientific) according to the manufacturer’s protocol. Three individual clones were fully sequenced with the sequencing primer shown in the Table 1.
Table 1: cing primers used to sequence the cytochrome P450-oxidoreductase cDNA M4 progeny of MCS 746 C.
SEQ ID NO Oligonucleotides sequence (5’ to 3’) pJET1.2 Forward CGACTCACTATAGGGAGAGCGGC Sequencing primer (SEQID NO 37) pJET1.2 e AAGAACATCGATTTTCCATGGCAG Sequencing primer (SEQID NO 38) Primer 1 (SEQ ID GCAGTGCTTCATATTTCTCG NO 39) Primer 2 (SEQ ID CCTTATGGAAAATATTGGAGGGAGC NO 40) Primer 3 (SEQ ID GTCTCTCTCAGAGGCTGCT NO 41) Primer 4(SEQ ID ACATCCAGGCAATC RNA-seq of stems and capsules from GSK NOSCAPINE CVS 3.
Stern and e material was harvested from stems and capsules of GSK NOSCAPINE CVS 3 plants 1-2 days after petal fall. RNA was ed individually from five plants. The harvested material was ground in liquid nitrogen using a mortar and pestle. RNA was ed from the ground stem or capsule preparations as previously described (Chang et al., 1993, Plant Mol. Biol. Rep. 11: 113-116) with slight modifications (three extractions with chloroform:isoamylalcohol, RNA precipitation with Lithium chloride at -20°C overnight). RNA was quantified spectrophotometrically before pooling equal amounts of RNA from five plants per cultivar, stage and organ. The pooled samples undenNent a final purification step using an RNeasy Plus MicroKit (Qiagen, Crawley, UK) to remove any remaining genomic DNA from the preparations. RNA was eluted in 100 uL RNase-free water.
RNA sequencing was performed on one lane of the lllumina HiSeq 2000 platform (lllumina, Little Chesterford, UK) using the TruSeq RNA kit. TruSeq RNA library construction was carried out according to the manufacturer’s description and included a poly-A selection step.
De novo sequence assembly of the raw ce datasets was performed using the Trinity RNA-seq de novo assembly tool (Haas et al., 2013, Nature Protocols, 8: 1494— 1512; http://trinityrnaseq.github.io/). on discovery in the STORR locus The clonal STORR cDNA ces obtained from M4 progeny of plant MCS 746 C displaying the low morphinan id/high noscapine phenotype were compared to the ponding contiguous sequence assembled from RNA-seq data from GSK NOSCAPINE CVS 3, the opium poppy ar used for EMA mutagenesis. The clonal and contiguous sequences were identical apart from a C to T transitions at position 1291 with respect to SEQ ID NO: 6. This C to T transition is consistent with the type of ons introduced by EMS e et al., 2003, Genetics, 164: 731-740). The mutation results in a arginine to phan amino acid substitution at position 431 of the STORR protein (R431VV).
Enzyme assays General reaction set up was carried out as previously described (Lenz & Zenk, 1995, FEBS 233:132—139). Substrates (S)-reticuline, 1,2—dehydroreticuline and coclaurine were purchased from TRC Chemicals (Toronto, Canada) and one was supplied by MacFarlan-Smith (Edinburgh, UK). Assay reactions contained 300 mM NADPH, 5 ug oxidoreductase A or B preparation, 100 mM buffer covering a range of pH values (potassium phosphate pH 6-8, glycine-NaOH pH 9) and 75 uM substrate. Reactions were incubated for 2 h at 37°C and immediately frozen at -80°C. Reactions were dried down to powder in speedvac Gene Vac EZ—2 plus (Ipswich, UK) at 40°C and ended in 100 uL 1:1 Hexane:Ethanol (v/v), 0.1% Diethylamine (v/v).
Alternatively enzyme assays were set up in 250 uL volumes containing final concentrations of 50 mM Bis-tris propane , 1 mM NADPH and 100 uM of substrate prepared from transformed yeast inantly expressing ype and mutant cytochrome P450—oxidoreductases. The assays were started by the addition of 50 uL of 2 mg mL'1 soluble or microsomal t from transformed yeast, and incubated at 37 °C for 3 hours. After this time, 50 uL of the extract was removed and quenched by the additional of 50 uL of methanol + 1 % acetic acid containing 100 mM noscapine as an internal rd. The reaction was then centrifuged at 20,000 x g for 2 min, and the supernatant recovered for analysis of 1,2-dehydroreticuline and (R/S) -reticuline content (i.e., iral method). The remaining 200 uL of assay was used for chiral is.
The reaction was quenched by the addition of 0.5 M sodium carbonated buffer (pH 10.0).
Reticuline was then extracted from the reaction products by three sequential extractions with 400 uL of dichloromethane. The solvent extracts were combined and evaporated to dryness in a speedvac. The extract was then dissolved in 100 uL of 50:50:0.1 hexane/ethanol/diethylamine and analysed by the chiral HPLC method to determine the relative amounts of (H)- and (S)-reticuline.
EMS nesis of a noscapine opium poppy cultivars leads to opium poppy lines where noscapine constitutes more than 90% of the total alkaloid t.
Among the plants derived from M2 seed line S—121435 one individual — designated plant A — displayed a high noscapine and low morphinan alkaloid phenotype compared to noscapine, thebaine and morphine cultivars (Table 3). The relative noscapine content expressed as percentage of total major alkaloids (morphine, codeine, ine, thebaine and noscapine) reached almost 85% in this plant. The relative content of noscapine further increased and that of morphinan alkaloids further decreased in the M3 and M4 progeny both in self-and ollinated capsules indicating that the underlying mutation is no longer segregating in these populations. Likewise, noscapine amounts to more than 90% of the relative alkaloid content in the latex of glasshouse-grown M4 siblings of plants used for sequencing the cytochrome P450-oxidoreductase (STORR) cDNA (Table 2). This shows that the mutant phenotype is stable in the M4 generation and across nmental and developmental conditions.
Generation of F2 populations population segregating storr-4 in the background of high noscapine cvs 4 (HN4) The impact of the storr—4 allele on noscapine and nan alkaloid accumulation was assessed in segregating F2 populations. M4 mutant plants derived from seed batch S- 186668 were crossed with high NOSCAPINE CVS4 (HN4). The resulting F1 tion was self-pollinated to produce F2 seed lines ating storr—4 in a noscapine background. Three F2 lines (S-185973, S-185975, S-185976) were grown along HN4 control plants in the glasshouse to maturity. Dried e material was harvested and analysed as described below.
Genotyping of storr-4 KASP genotyping assays for storr—4 were designed to genotype F2 populations. To design allele-specific primers, sequences of 50-100 nucleotides around the mutation site were submitted to LGC genomics (Hoddesdon, UK) to order KASP by Design (KBD) primer mix. The sequences of the allele-specific primers as well as the common primer are shown in Table 4.
Table 4. Primer seo uences, in 5’-3’ orientation.
Mutant Primer Primer sequence Label allele "M type a"e'e CCAGGCAATCATCAAAGAATCAATGC FAM (SEQ ID NO 34) storr—4 Mutant allele CCAGGCAATCATCAAAGAATCAATGT SEQ ID NO 35 Common primer CACGGGGCTGGCTGGATACAA - (SEQ ID NO 36) Leaf samples (30-50mg) for DNA extraction were harvested from 4-6 week old plants.
Genomic DNA was extracted using the BioSprint 96 Plant kit on the BioSprint 96 Workstation (Qiagen, y, UK) ing to the manufacturer’s ol. Extracted DNA was quantified on the opT'VI 8000 Spectrophotometer (Fisher Scientific, VWmington, DE, USA) and normalized to 5 ng/uL.
KASP assay reactions were carried out in FrameStar® 384 well plates (4titude® Ltd, Wotton, UK) in a 5.07 uL volume per reaction, with the following reaction components: 2x KASP V4.0 mix (LGC genomics), 2.5 uL; KBD primer mix, 0.07 uL; HyClone molecular grade water (Thermo ific, Hemel Hempstead, UK), 0.5 uL; 5 ng/uL DNA, 2 uL. The plates were centrifuged for 2 minutes at 4500 x g, heat-sealed using the KubeTM heat-based plate sealer (LGC genomics), and thermal cycling was carried out in a HydrocyclerTM (LGC genomics) water bath-based thermal cycler, with the following conditions: 94°C for 15 minutes; 10 cycles: 94°C for 20 seconds, 61-55°C for 60 seconds (dropping 06°C per cycle); 34 cycles: 94°C for 20 seconds, 55°C for 60 seconds. The plate reading was carried out on an d Biosystems ViiA7 instrument (Life Technologies), for 30 seconds at 25°C.
Capsule straw analysis of glasshouse grown F2 populations es from the F2 populations and HN4 controlswere ted once capsules had dried to approximately 10% moisture on the plants. Seed was manually separated from the e and single capsules were ground to a fine powder in a ball mill (Model MM04, , Haan, Germany). Samples of ground poppy straw were then weighed accurately to 10 1r 0.1 mg and extracted in 0.5 mL of a 10% (v/v) acetic acid on with gentle shaking for 1h at room ature. Samples were then clarified by centrifugation and a 50 uL subsample diluted 10x in 1% (v/v) acetic acid to give an alkaloid solution in 2% acetic acid for further is. All solutions were ed as described for the poppy straw analysis from field grown plants. Likewise, all data analysis was carried out using the R programming language. Putative alkaloid peaks were quantified by their pseudomolecular ion areas using custom scripts. Alkaloids were identified by comparing exact mass and retention time values to those of standards. Where standards were not available, the Bioconductor rcdk e (Smith et al., 2006), Anal. Chem. 78 (3): 779- 787) was used to generate pseudomolecular formulae from exact masses within elemental constraints C = 1 100, H = 1 200, O = 0 200, N = 0 3 and mass accuracy < 5ppm. The hit with the lowest ppm error within these constraints was used to assign a putative formula.
Examples The storr-4 allele co-segregates with the high noscapine zero morphinan phenotype.
F2 populations segregating storr—4 in a high ine background were genotyped for storr—4 and their relative and absolute alkaloid content determined in dried capsules. The storr—4 mutant allele strictly co-segregated with extremely high noscapine and zero morphinan levels (Figure 1 and 2). The relative noscapine content in homozygous storr—4 al reached 98 :r 0.7 % with the remainder being mainly reticuline (1.8 :r 0.7 %).
Noscapine and morphinan levels of zygous F2 material were intermediate between those of homozygous storr—4 F2 material and F2 plants gous for the STORR wild type allele (Figure 1 and 2). The alkaloid profile of F2 plants homozygous for the STORR wild type allele was overall very similar to that of control plants from HN4, one of the parental ies used to te the F2 population.
The data clearly demonstrate that the shutdown of morphinan biosynthesis in STORR knock out mutations such as 4 allows leads to increased metabolic flux into the noscapine branch pathway (Figure 3) thus resulting in ely high levels of noscapine.
Table 2 iLatex data from glasshouse—grown M4 progeny (5—186668) of plant MCS 746 C Alkaloid content % total alkaloids Morphine Codeine Codeinone Oripavine Thebaine Noscapine Laudanosin Protopine 1.3 i 2.32 0.34 i 0.71 0.01 i 0 0.07 i 0.1 .2.34 i 3.3' 0.81 i 0.38 0.2 i 0.22 fiaverages and standard deviation of 15 individual M4 plants are shown Table 3 alkaloid content (% dry weight and % total nd content) in different plant lines 38.8w :3 **%_o_e__~ mcfimumozgfinmsh HRS :md .33 W x 9.33:0 . 23:8 W 229.2 mEmuou H84 #3 Hmmmd Nde ." HR "mod m3 38.0 ~de E225 :Emumozgfinmf. +mmoomI. Sod 23:8 9.33:0 mo+NooI.
W 05380: 229:4. mEmuou m+mmooI. mHod vcm 9:59.05. +m~ooI wEmnwE. . mood \Amvflsmamu 552320 53:39 3.35ch.. o-< o-< 8:8 38 vamE am am £5208 Mcozfimcmow 09:3 mw>uNzau :6 H N m>u Eamudm £55822 I m5 m>u ms: z&o_>_ mz_I&o_>_ Emmee 388 mz_833 2338 MEoEvzm 9K sz_ _0Q EmE View v7.9 mmENTWW View v_m View Us: mmENHh vamchawEM View View 92W "am 335209 :5 :30 co commmm mH\NHo~ 358 338 u no vwmmn

Claims (65)

Claims
1. A plant of the genus Papaver wherein said plant is modified in a gene encoding a cytochrome P450-oxidoreductase catalysing the conversion of (S)-reticuline to (R)-reticuline wherein said modification is stably inherited and s or abrogates the sion or activity of said cytochrome P450 domain and/or oxidoreductase domain of said modified polypeptide compared to a ype plant comprising a functional cytochrome P450- oxidoreductase, wherein said plant comprises a noscapine content of between at least 55 to at least 99% of the total content of morphine, codeine, oripavine, ne and noscapine, and between 1.8 and 5% ine of capsule dry weight.
2. The plant according to claim 1 wherein said plant is modified in a gene comprising a nucleotide sequence selected from the group consisting of: i) a nucleic acid molecule comprising a tide sequence as set forth in SEQ ID NO: 1 or 2 or 3; ii) a nucleic acid molecule comprising a nucleotide sequence as set forth in SEQ ID NO: 4 or 33; iii) a nucleic acid molecule comprising a nucleotide sequence as set forth in SEQ ID NO: 5; iv) a nucleic acid molecule comprising a nucleic acid sequence as set forth in SEQ ID NO: 6, 7 or 8; v) a c acid molecule the complementary strand of which hybridizes under ent hybridization conditions to the nucleotide sequence set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33 and which encodes a polypeptide that has reduced or abrogated expression or activity of a cytochrome P450-oxidoreductase catalysing the conversion of (S)- line to (R)-reticuline compared to a wild-type Papaver plant comprising a gene ng a functional cytochrome P450- oxidoreductase.
3. The plant according to claim 1 or 2 wherein said modified nucleic acid molecule is at least 50% identical to the nucleotide sequence set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or
4. The plant according to claim 3 wherein said ed nucleic acid molecule is at least 55%, 60%, 65%, 70%, 75%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the nucleotide sequence set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33 over the full nucleotide sequence.
5. The plant according to claim 4 wherein said modified c acid molecule is at least 99% identical to the nucleotide ce set forth in SEQ ID NO 6, 7 or 8, over the full nucleotide sequence and wherein said modification is an addition, substitution or deletion of at least one nucleotide base between tides 1280-1300 set forth in SEQ ID NO 6, 7 or
6. The plant according to claim 5 wherein said modification is a substitution of at least one nucleotide base n nucleotides 1280-1300 set forth in SEQ ID NO 6, 7 or 8.
7. The plant according to claim 6 wherein said modification is a tution of at least one nucleotide base between nucleotides 1285-1295 set forth in SEQ ID NO 6, 7 or 8.
8. The plant according to claim 7 n said modification is a C to T substitution of the nucleotide base at nucleotide 1291 of SEQ ID NO 6 or 7 or at nucleotide 1294 of SEQ ID NO:
9. The plant according to any one of claims 1 to 8 wherein said plant expresses a modified P450-oxidoreductase polypeptide wherein said modified polypeptide has reduced or ted activity in the conversion of (S)-reticuline to (R)-reticuline.
10. The plant according to claim 9 wherein said ed polypeptide is selected from the group ting of: i) a polypeptide comprising a modified amino acid sequence of the amino acid sequence set forth in SEQ ID NO: 9, 10 or 11; ii) a polypeptide comprising a modified amino acid sequence of the amino acid sequence as set forth in SEQ ID NO: 12, 13 or 14; iii) a polypeptide comprising a modified amino acid sequence that is at least 95% identical to the full length amino acid sequence as set forth in SEQ ID NO: 9, 10, 11, 12, 13 or 14, wherein said sequence is modified by addition, deletion or tution of one or more amino acid sequences and wherein said polypeptide has reduced or abrogated enzyme activity in the conversion of (S)-reticuline to 1,2- dehydroreticuline and/or to (R)-reticuline when compared to the unmodified polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 9, 10, 11, 12, 13 or 14.
11. The plant according to claim 9 wherein said modified polypeptide is selected from the group consisting of: i) a modified polypeptide comprising a ed amino acid sequence of the amino acid sequence set forth in SEQ ID NO: 15; ii) a modified polypeptide sing a modified amino acid sequence of the amino acid sequence set forth in SEQ ID NO: 16; iii) a modified polypeptide comprising a modified amino acid sequence that is at least 95% identical to the full length amino acid sequence as set forth in SEQ ID NO: 15 or 16, wherein said sequence is modified by addition, deletion or tution of one or more amino acid sequences and wherein said polypeptide has reduced or abrogated enzyme activity in the conversion of 1,2-dehydroreticuline to (R)-reticuline when ed to the unmodified polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 15 or 16.
12. The plant ing to claim 9 or 10 wherein said modified polypeptide comprises at least an addition, deletion or substitution of an amino acid residue in the region of amino acids 386-486 as set forth in SEQ ID NO: 9, 10 or 11.
13. The plant according to claim 12 wherein said modified polypeptide comprises at least an addition, deletion or substitution of an amino acid residue in the region of amino acids 426- 436 as set forth in SEQ ID NO: 9, 10 or 11 and wherein said modified polypeptide is at least 90% identical to the amino acid sequence set forth in SEQ ID NO: 9, 10 or 11.
14. The plant according to any one of claims 9 or 10 wherein said modified ptide comprises a deletion or substitution of amino acid residue 431 of SEQ ID NO: 9 wherein said modified polypeptide is at least 90% identical to the amino acid sequence set forth in SEQ ID NO: 9.
15. The plant according to claim 9 or 10 to 13 wherein said modified polypeptide comprises a on or substitution of amino acid residue 432 of SEQ ID NO: 10 wherein said modified polypeptide is at least 90% identical to the amino acid sequence set forth in SEQ ID NO: 10.
16. The plant according to claim 9 or 10 wherein said modified polypeptide comprises a deletion or substitution of amino acid residue 431 of SEQ ID NO: 11 wherein said modified polypeptide is at least 90% cal to the amino acid sequence set forth in SEQ ID NO: 11.
17. The plant according to claim 9 or 10 wherein said ed polypeptide comprises a substitution or deletion at amino acid arginine 431 of the amino acid sequence set forth in SEQ ID NO: 9.
18. The plant according to claim 9 or 10 wherein said modified polypeptide comprises a substitution or deletion at amino acid arginine 432 of the amino acid sequence set forth in SEQ ID NO: 10.
19. The plant according to claim 9 or 10 wherein said modified polypeptide comprises a substitution or deletion at amino acid arginine 431 of the amino acid sequence set forth in SEQ ID NO: 11.
20. The plant ing to claim 9 or 10 wherein said modification is the substitution of arginine for phan in at position 431 of the amino acid sequences set forth in SEQ ID NO: 9.
21. The plant according to claim 9 or 10 n said modification is the substitution of ne for tryptophan in at position 432 of the amino acid sequences set forth in SEQ ID NO: 10.
22. The plant according to claim 9 or 10 wherein said modification is the substitution of arginine for tryptophan in at position 431 of the amino acid sequences set forth in SEQ ID NO: 11.
23. The plant according to claim 9 or 10 wherein said modified polypeptide comprises the amino acid sequence set forth in SEQ ID NO: 27.
24. The plant according to claim 9 or 10 wherein said modified polypeptide comprises the amino acid sequence set forth in SEQ ID NO: 28.
25. The plant according to claim 9 or 10 wherein said modified ptide comprises the amino acid sequence set forth in SEQ ID NO: 29.
26. The plant according to claim 9 or 10 wherein said modified polypeptide is encoded by a nucleic acid molecule sing a nucleotide sequence set forth in SEQ ID NO: 30.
27. The plant according to claim 9 or 10 wherein said modified polypeptide is encoded by a nucleic acid molecule comprising a nucleotide sequence set forth in SEQ ID NO: 31.
28. The plant according to claim 9 or 10 wherein said modified polypeptide is encoded by a nucleic acid molecule comprising a nucleotide sequence set forth in SEQ ID NO: 32.
29. The plant according to any one of claims 1 to 28 wherein said plant is zygous for said modified gene.
30. The plant according to any one of claims 1 to 28 wherein said plant is homozygous for said modified gene.
31. The plant according to any one of claims 1 to 30 n said plant comprises one or more genes comprising a nucleotide sequence selected from the group consisting of: i) a nucleotide sequence as represented by the sequence in SEQ ID NO: 17,18, 19, 20, 21, 22, 23, 24, 25 or 26; ii) a nucleotide ce wherein said sequence is degenerate as a result of the genetic code to the nucleotide ce defined in (i); iii) a nucleic acid le the complementary strand of which hybridizes under ent hybridization conditions to the sequence in SEQ ID NO: 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 wherein said nucleic acid molecule encodes polypeptides involved in the biosynthesis of Papaver noscapine or ediates in the biosynthesis of noscapine.
32. The plant according to claim 31 wherein said plant comprises enhanced levels of noscapine and intermediates in the synthesis of noscapine.
33. The plant according to claim 32 wherein said plant has a noscapine content of between 2.55% - 5% dry weight.
34. The plant according to claim 32 wherein said plant has a noscapine content of between 2.55% - 4.22% dry weight.
35. The plant according to claim 32 wherein said plant has a noscapine content of between 3.37% - 4.22% dry .
36. The plant according to claim 32 wherein noscapine comprises at least 60% of the benzylisoquinoline alkaloid content of said ed plant.
37. The plant according to claim 33 or 34 wherein noscapine comprises at least 99% of the benzylisoquinoline content of said modified plant
38. The plant according to any one of claims 1 to 37 wherein said Papaver plant is r somniferum.
39. A Papaver plant according to claim 1, wherein said plant is modified by transformation or transfection with an expression vector adapted to express an antisense, siRNA or shRNA targeting SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33 wherein said modified plant comprises a noscapine content of between at least 55 to at least 99% of the total content of ne, e, oripavine, thebaine and noscapine, and between 1.8 and 5% ine of capsule dry weight.
40. The plant according to claim 39 wherein said modified plant comprises a noscapine content that is at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% of the of the benzylisoquinoline alkaloid content of said modified plant.
41. The plant according to claim 39 or 40 wherein said modified plant comprises a benzylisoquinoline alkaloid content that comprises substantially noscapine.
42. A Papaver capsule obtained from the modified plant according to any one of claims 1 to 41.
43. A Papaver straw obtained from the modified plant according to any one of claims 1 to
44. A Papaver seed obtained from the modified plant according to any one of claims 1 to
45. A modified ptide comprising the amino acid sequence set forth in SEQ ID NO:
46. A modified polypeptide comprising the amino acid sequence set forth in SEQ ID NO:
47. A modified polypeptide comprising the amino acid sequence set forth in SEQ ID NO:
48. A nucleic acid molecule comprising the nucleotide sequence set forth in SEQ ID NO:
49. A nucleic acid molecule comprising the tide ce set forth in SEQ ID NO:
50. A nucleic acid le comprising the nucleotide sequence set forth in SEQ ID NO:
51. A method to obtain a Papaver plant comprising enhanced or elevated noscapine, or intermediates in the synthesis of noscapine, n said plant is modified in a gene encoding a cytochrome xidoreductase catalysing the conversion of ticuline to (R)-reticuline wherein said modification is stably inherited and reduces or abrogates the expression or activity of said rome P450 domain and/or oxidoreductase domain of said modified polypeptide compared to a ype plant comprising a functional cytochrome P450- oxidoreductase, wherein said plant comprises a noscapine content of between at least 55 to at least 99% of the total content of morphine, codeine, oripavine, thebaine and ine, and between 1.8 and 5% ine of capsule dry weight, said method comprising the steps i) mutagenesis of wild-type seed from a Papaver plant that comprises a gene encoding a functional cytochrome xidoreductase catalysing the conversion of (S)-reticuline to (R)-reticuline; ii) cultivation of the seed in i) to produce first and subsequent generations of iii) obtaining seed from the first generation plant and subsequent generations of plants; iv) ining if the seed from said first and subsequent generations of plants has altered nucleotide sequence of said encoding a cytochrome P450- oxidoreductase; v) obtaining a sample and analysing the nucleic acid sequence of a nucleic acid molecule selected from the group consisting of: i. a nucleic acid molecule comprising a nucleotide sequence as represented in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33; ii. a nucleic acid le that hybridises to a complementary strand of said nucleic acid molecule in i) under stringent hybridisation conditions and that encodes a non-functional cytochrome P450- oxidoreductase; and optionally vi) comparing the nucleotide sequence of the nucleic acid molecule in said sample to a nucleotide sequence of a nucleic acid molecule of the al wild-type plant.
52. The method according to claim 51 wherein said plants have an altered expression of said cytochrome P450-oxidoreductase.
53. The method ing to claim 51 or 52 wherein said modified nucleic acid molecule is at least 55%, 60%, 65%, 70%, 75%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the nucleotide sequence set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33 over the full nucleotide sequence.
54. The method according to any one of claims 51 to 53 wherein said nucleic acid molecule is analysed by a method comprising the steps of: i) extracting c acid from said d plants; ii) amplification of a part of said nucleic acid molecule by a polymerase chain reaction; iii) g a preparation comprising the amplified nucleic acid and nucleic acid extracted from wild-type seed to form heteroduplex nucleic acid; iv) incubating said preparation with a single stranded nuclease that cuts at a region of heteroduplex c acid to identify the mismatch in said heteroduplex; and v) determining the site of the mismatch in said nucleic acid duplex.
55. The method according to any one of claims 51 to 54 wherein said modified plant is analysed for noscapine content and optionally compared to the original unmodified plant.
56. A plant obtained by the method according to any one of claims 1 to 41 and 51 to 55.
57. The use of a gene encoding a cytochrome P450-oxidoreductase catalysing the sion of (S)-reticuline to (R)-reticuline in the identification of Papaver cultivars that have enhanced levels of noscapine or intermediates in noscapine synthesis, wherein said cultivars comprise a modified gene encoding a cytochrome P450-oxidoreductase catalysing the conversion of (S)-reticuline to (R)-reticuline wherein said modification is stably inherited and reduces or abrogates the expression or activity of said rome P450 domain and/or oxidoreductase domain of said modified polypeptide compared to a wild-type plant comprising a functional cytochrome P450-oxidoreductase, wherein said cultivar comprises a noscapine content of between at least 55 to at least 99% of the total content of morphine, codeine, oripavine, thebaine and noscapine, and n 1.8 and 5% noscapine of e dry weight, in a method comprising the steps: i) obtaining a sample of plant material from a plant to be tested; ii) extracting genomic nucleic acid from said plant material; and iii) analysing the extracted c nucleic acid encoding a variant nucleotide sequence of the tide ces set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33; and optionally, iv) comparing the ce to a nucleotide sequence sing SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33.
58. Use according to claim 57 wherein said gene comprises a nucleotide sequence selected from the group consisting of: i) a nucleic acid molecule sing a nucleotide sequence as set forth in SEQ ID NO: 1, 2 or 3; ii) a nucleic acid molecule molecule comprising a nucleotide sequence as set forth in SEQ ID NO: 4 or 33; iii) a nucleic acid molecule molecule sing a nucleotide sequence as set forth in SEQ ID NO: 5; iv) a nucleic acid molecule molecule comprising a nucleic acid sequence as set forth in SEQ ID NO: 6, 7 or 8; v) a nucleic acid molecule the complementary strand of which hybridizes under stringent hybridization conditions to the nucleotide sequence set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33 and which encodes a polypeptide that has reduced or abrogated expression or activity of a cytochrome P450- eductase catalysing the conversion of (S)-reticuline to ticuline ed to a wild-type Papaver plant comprising a gene encoding a functional cytochrome P450-oxidoreductase.
59. A method to identify a Papaver plant comprising a ed cytochrome P450- oxidoreductase gene, wherein said plant is modified in a gene encoding a cytochrome P450- oxidoreductase catalysing the conversion of (S)-reticuline to (R)-reticuline wherein said modification is stably inherited and reduces or abrogates the expression or activity of said cytochrome P450 domain and/or oxidoreductase domain of said modified polypeptide ed to a wild-type plant comprising a functional cytochrome P450-oxidoreductase, wherein said plant comprises a noscapine content of between at least 55 to at least 99% of the total t of morphine, codeine, oripavine, thebaine and noscapine, and between 1.8 and 5% noscapine of capsule dry weight, said method comprising the steps: i) obtaining a sample of plant material from a plant to be ; ii) extracting genomic nucleic acid from said plant material; and iii) analysing the extracted genomic nucleic acid encoding a variant nucleotide sequence of the nucleotide sequences set forth in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33; and optionally, iv) ing the sequence to a nucleotide sequence comprising SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8 or 33.
60. A method according to claim 59 wherein said analysis comprises the steps: i) preparing a reaction mixture comprising: a) genomic DNA to be analysed, b) a first allele specific forward oligonucleotide primer comprising a nucleotide sequence complementary to said cytochrome P450-oxidoreductase gene nucleotide sequence and a nucleotide sequence complementary to a nucleotide sequence of a first fluorescent reporter cassette, c) a second allele specific forward oligonucleotide primer comprising a nucleotide ce complementary to said cytochrome P450-oxidoreductase gene an a nucleotide sequence complementary to a nucleotide sequence of a second fluorescent reporter cassette wherein the nucleotide at the 3’-end of said second oligonucleotide primer ce is complimentary to a polymorphic nucleotide sequence variant of said rome xidoreductase gene, and wherein the nucleotide sequence of said first and second oligonucleotide primers are complementary to the same upstream nucleotide sequence of said cytochrome P450- oxidoreductase gene; d) a reverse oligonucleotide primer comprising a nucleotide sequence complementary to a nucleotide sequence downstream of said cytochrome P450- oxidoreductase gene nucleotide sequence; and e) two reporter cassettes comprising double stranded nucleic acid comprising: a first double stranded nucleic acid wherein one strand comprises a scent label and is complementary to said first allele specific primer and a second strand comprising a quencher label that quenches said fluorescent label; and a second double ed nucleic acid wherein one strand comprises a scent label ent from the fluorescent label of i) and is complementary to said second allele specific primer and a second strand comprising a quencher label that quenches said second scent label; ii) g said reaction mixture to denature genomic nucleic acid and said double stranded reporters and providing conditions to anneal primers to their tive complementary nucleotide sequence, primer extending said annealed oligonucleotide primers and polymerase chain ication of said mixture; and iii) detecting fluorescence emission.
61. A method according to claim 60 wherein said modified and wild type cytochrome P450-oxidoreducatse gene nucleic acid sequence is as set forth SEQ ID NO: 6.
62. A method according to claims 60 or 61 wherein said first (forward) primer comprise (SEQ ID NO: 35) AATCATCAAAGAATCAATGT, said second rd) primer comprises (SEQ ID NO: 34) CCAGGCAATCATCAAAGAATCAATGC and said reverse primer comprises (SEQ ID NO 36) CACGGGGCTGGCTGGATACAA.
63. The method or use according to any one of claims 60 to 62 wherein said use or method includes analysis of the sequence or expression of a further gene selected from the group consisting of: i) a tide sequence as represented by the sequence in SEQ ID NO: 17, 18, 19 20, 21, 22, 23, 24, 25 or 26; ii) a nucleotide sequence wherein said sequence is rate as a result of the genetic code to the nucleotide sequence d in (i); iii) a nucleic acid molecule the complementary strand of which hybridizes under stringent hybridization conditions to the sequence in SEQ ID NO: 17, 18, 19 20, 21, 22, 23, 24, 25 or 26 wherein said nucleic acid molecule encodes ptides involved in the biosynthesis of Papaver noscapine or intermediates in the biosynthesis of noscapine.
64. A method for the extraction of noscapine from a modified plant or capsule or straw according to the invention comprising the steps: i) ting a modified plant, capsule or straw according any one of claims 1 to 43 and forming a reaction mixture of particulate plant al; ii) solvent extracting said reaction e to provide an alkaloid enriched fraction; and iii) concentrating said enriched alkaloid fraction to provide an enriched noscapine fraction.
65. A plant according to claim 1, wherein said plant comprises a noscapine content of at least 70% of the total content of morphine, codeine, oripavine, thebaine and noscapine, and over 2.55% noscapine of the capsule dry weight.
NZ738299A 2015-06-24 2016-06-23 Production of noscapine NZ738299B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB1511156.0A GB201511156D0 (en) 2015-06-24 2015-06-24 Production of alkaloids
GB1511156.0 2015-06-24
PCT/GB2016/051887 WO2016207643A1 (en) 2015-06-24 2016-06-23 Production of noscapine

Publications (2)

Publication Number Publication Date
NZ738299A NZ738299A (en) 2022-03-25
NZ738299B2 true NZ738299B2 (en) 2022-06-28

Family

ID=

Similar Documents

Publication Publication Date Title
AU2018101471A4 (en) Production of noscapine
Gu et al. An-2 encodes a cytokinin synthesis enzyme that regulates awn length and grain production in rice
Cotsaftis et al. Root-specific transcript profiling of contrasting rice genotypes in response to salinity stress
Brutnell et al. BUNDLE SHEATH DEFECTIVE2, a novel protein required for post-translational regulation of the rbcL gene of maize
US9862979B2 (en) Biosynthesis of opiate alkaloids
US10385354B2 (en) Papaver somniferum cytochrome P450
Yang et al. Development of AS‐PCR marker based on a key mutation confirmed by resequencing of W x‐mp in Milky P rincess and its application in japonica soft rice (O ryza sativa L.) breeding
US9458481B2 (en) Methyltransferase nucleic acids and polypeptides
WO2010147196A1 (en) Protein having novel prenylation enzyme activity and gene encoding same
Lee et al. Exploiting expressed sequence tag databases for the development and characterization of gene‐derived simple sequence repeat markers in the opium poppy (Papaver somniferum L.) for forensic applications
WO2021195780A1 (en) Methods of determining sensitivity to photoperiod in cannabis
AU2021102558A4 (en) Modified plant
NZ738299B2 (en) Production of noscapine
KR102154701B1 (en) Novel genetic markers for selection of watermelon dwarf entities and use thereof
KR102584678B1 (en) ‘Samkwang(SA)-flo3’, floury endosperm rice mutant derived from ‘Samkwang’
KR101500613B1 (en) Discrimination method for rice genetic resource and primers for discrimination of rice having amylose content
Armoniene et al. Nonsense-mediated decay of sucrose synthase 1 mRNA with induced premature chain termination codon during cold acclimation in winter wheat
US12002546B2 (en) Methods of determining sensitivity to photoperiod in cannabis
JPH0568564A (en) Rice plant peroxidase gene
Vrbovský et al. New Low Morphine Opium Poppy Genotype Obtained by TILLING Approach. Plants 2023, 12, 1077
KR20170086171A (en) Molecular marker for determining yellow fruit in tomato and sorting method for yellow tomato using same
WO2011108104A1 (en) Method of strengthening seed dormancy of wheat by accumulating gene mutations encoding abscisic acid metabolizing enzymes
WO2024129991A1 (en) Methods for producing soybean with altered composition
Frerichmann et al. Detection of new sequence variants for flowering and bolting time genes in species of the genus Beta