Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL POLYPEPTIDES FOR PRODUCING ALBICANOL AND/OR DRIMENOL COMPOUNDS
Document Type and Number:
WIPO Patent Application WO/2021/105236
Kind Code:
A2
Abstract:
The present invention relates to novel plant-derived haloacid dehalogenase-like (HAD-like) polypeptides having cyclic terpene synthase (TPS) activity, in particular suitable for use in biochemical methods of producing drimane-type sesquiterpenes, encompassing drimenol and/or albicanol and related compounds, like phosphorylated drimane-type sesquiterpene alcohols, in particular phosphorylated drimenol and/or albicanol compounds and derivatives. The invention also provides the coding nucleotide sequences of said novel TPS activity, corresponding expression constructs, recombinant hosts, method of preparing such novel polypeptides and mutants and variants thereof. The invention also relates to the use of such novel polypeptides in the production of odorants, flavours and fragrance ingredients.

Inventors:
WANG QI (CN)
SCHALK MICHEL (CH)
Application Number:
PCT/EP2020/083408
Publication Date:
June 03, 2021
Filing Date:
November 25, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
FIRMENICH & CIE (CH)
International Classes:
C12P5/00; C12N9/88; C12P7/02; C12P7/04
Domestic Patent References:
WO2018220113A12018-12-06
Foreign References:
EP1149849A12001-10-31
EP1069183A22001-01-17
DE10019373A12001-10-31
EP2019063824W2019-05-28
Other References:
TETRAHEDRON: ASYMMETRY, vol. 11, 2000, pages 1375 - 1388
PEARSONLIPMAN, PROC. NATL. ACAD, SCI. (USA, vol. 85, no. 8, 1988, pages 2444 - 2448
A. BATEMANE. BIRNEYL. CERRUTIR. DURBINL. ETWILLERS. R. EDDYS. GRIFFITHS-JONESK. L. HOWEM. MARSHALLE. L. L. SONNHAMMER, NUCLEIC ACIDS RESEARCH, vol. 30, no. 1, 2002, pages 276 - 280
J. LALONDEA. MARGOLINK. DRAUZH. WALDMANN: "Enzyme Catalysis in Organic Synthesis", vol. III, 2002, COLD SPRING HARBOR LAB PRESS, article "Immobilization of Enzymes", pages: 991 - 1032
TATIANA ET AL., FEMS MICROBIOL LETT., vol. 174, 1999, pages 247 - 250
BARIK S, MOL BIOTECHNOL, vol. 3, 1995, pages 6.3.1 - 6.3.6
SAMBROOK, J.FRITSCH, E.F.MANIATIS, T.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY, COLD SPRING HARBOR LABORATORY PRESS
SAMBROOKRUSSELL: "Molecular Cloning", 2001, COLD SPRING HARBOR LABORATORY PRESS
GREENER ACALLAHAN MJERPSETH B: "vitro mutagenesis protocols", 1996, HUMANA PRESS, article "An efficient random mutagenesis technique using an E.coli mutator strain"
BARETTINO DFEIGENBUTZ MVALCAREL RSTUNNENBERG HG, NUCLEIC ACIDS RES, vol. 22, 1994, pages 1593
ECKERT KAKUNKEL TA, NUCLEIC ACIDS RES, vol. 18, 1990, pages 3739
SCHENK ET AL., BIOSPEKTRUM, vol. 3, 2006, pages 277 - 279
STEMMER WPC, NATURE, vol. 370, 1994, pages 389
STEMMER WPC, PROC NATL ACAD SCI USA, vol. 91, 1994, pages 10747
REETZ MTJAEGER K-E, TOPICS CURR CHEM, vol. 200, 1999, pages 31
ZHAO HMOORE JCVOLKOV AAARNOLD FH: "Manual of industrial microbiology and biotechnology, American Society for Microbiology", 1999, article "Methods for optimizing industrial polypeptides by directed evolution"
GOEDDEL: "Gene Expression Technology: Methods in Enzymology", vol. 185, 1990, ACADEMIC PRESS
"Cloning Vectors", 1985, ELSEVIER
T.J. SILHAVYM.L. BERMANL.W. ENQUIST: "Biotechnologie - Lehrbuch der angewandten Mikrobiologie", 1984, COLD SPRING HARBOR LABORATORY
AUSUBEL, F.M. ET AL.: "Applied Microbiol. Physiology, A Practical Approach", 1997, GREENE PUBLISHING ASSOC. AND WILEY INTERSCIENCE, pages: 53 - 73
PATEK ET AL., APPL. ENVIRON. MICROBIOL., vol. 60, 1994, pages 133 - 140
"Manual of Methods for General Bacteriology", 1981, AMERICAN SOCIETY FOR BACTERIOLOGY
HARLOW, E.LANE, D.: "Biochemische Arbeitsmethoden [Biochemical processes", 1988, COLD SPRING HARBOR (N.Y.) PRESS
MALAKHOVA ET AL., BIOTEKHNOLOGIYA, vol. 11, 1996, pages 27 - 32
SCHMIDT ET AL., BIOPROCESS ENGINEER, vol. 19, 1998, pages 67 - 70
"Ullmann's Encyclopedia of Industrial Chemistry", vol. A27, 1996, VCH: WEINHEIM, pages: 89 - 90,521-540,540-547,559-566,575-581,581-587
MICHAL, G: "Biochemical Pathways: An Atlas of Biochemistry and Molecular Biology", 1999, JOHN WILEY AND SONS
FALLON, A. ET AL.: "Applications of HPLC in Biochemistry", LABORATORY TECHNIQUES IN BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 17, 1987
"Bioprocess technology 1. Introduction to bioprocess technology", CHMIEL: BIOPROZESSTECHNIK: EINFIIHRUNG IN DIE BIOVERFAHRENSTECHNIK, vol. 1
CHMIELHAMMESBAILEY, BIOCHEMICAL ENGINEERING
Attorney, Agent or Firm:
BAUMGARTNER HARRIS, Pauline (CH)
Download PDF:
Claims:
Claims

1. An isolated polypeptide of the haloacid dehalogenase-like (HAD-like) hydrolase superfamily, comprising cyclic terpene synthase activity, wherein said polypeptide is a. BazzHADl comprising an amino acid sequence of SEQ ID NO: 3, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40% sequence identity to SEQ ID NO: 3 and retaining said terpene synthase activity; b. BazzHAD2 comprising an amino acid sequence of SEQ ID NO: 6, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40% sequence identity to SEQ ID NO: 6 and retaining said terpene synthase activity; c. BazzHAD3 comprising an amino acid sequence of SEQ ID NO: 9, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40% sequence identity to SEQ ID NO: 9 and retaining said terpene synthase activity; d. BtHAD comprising an amino acid sequence of SEQ ID NO: 12, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40% sequence identity to SEQ ID NO: 12 and retaining said terpene synthase activity; e. SmHADl comprising an amino acid sequence of SEQ ID NO: 19, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40% sequence identity to SEQ ID NO: 19 and retaining said terpene synthase activity; f. SmHAD2 comprising an amino acid sequence of SEQ ID NO: 26, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40% sequence identity to SEQ ID NO: 26 and retaining said terpene synthase activity;

2. The polypeptide of claim 1, comprising the ability to produce a) a phosphate precursor of at least one drimane sesquiterpene; or b) at least one drimane sesquiterpene; or c) the phosphate precursor of at least one drimane sesquiterpene and the corresponding at least one drimane sesquiterpene.

3. The polypeptide of claim 1 or 2, comprising the ability to produce a) drimenyl diphosphate and/or albicanyl diphosphate from famesyl diphosphate (FPP) as substrate; or b) drimenol and/or albicanol from FPP as substrate, either directly from FPP as substrate or via the respective diphosphate precursor; or c) drimenyl diphosphate and/or albicanyl diphosphate from famesyl diphosphate (FPP) as substrate; and drimenol and/or albicanol, either directly from FPP as substrate or via the respective diphosphate precursor.

4. The polypeptides of anyone of claims 1 to 3 further comprising a. a modified class II terpene synthase motif as set forth in SEQ ID NO: 46 (PxoDxiD(T/S)(T/M)S), wherein xo and xi can be any naturally occurring amino acid residue, and xi particularly represents I or L, in particular the motif of any one of SEQ ID NOs: 47, 48, 49 or 50, wherein said modified class II terpene synthase motif corresponds to i. sequence positions 318 to 325 of SEQ ID NO: 3, ii. sequence positions 269 to 276 of SEQ ID NO: 6, iii. sequence positions 277 to 284 of SEQ ID NO: 9, iv. sequence positions 318 to 325 of SEQ ID NO: 12, v. sequence positions 255 to 262 of SEQ ID NO: 19, and vi. sequence positions 255 to 262 of SEQ ID NO: 26, respectively; b. a QW motif as set forth in SEQ ID NO: 51 (QX2X3DGX4W), wherein X2, X3 and X4 can be any naturally occurring amino acid residue, and X4 particularly represents G or S, in particular the motif of any one of SEQ ID NOs: 52, 53, 54, or 55, wherein said QW motif corresponds to i. sequence positions 489 to 495 of SEQ ID NO: 3, ii. sequence positions 431 to 437 of SEQ ID NO: 6, iii. sequence positions 439 to 445 of SEQ ID NO: 9, iv. sequence positions 488 to 494 of SEQ ID NO: 12, v. sequence positions 430 to 436 of SEQ ID NO: 19, and vi. sequence positions 430 to 436 of SEQ ID NO: 26, respectively; and c. optionally at least one further sequence motif selected from

(1) the conserved sequence motif 1 set forth in SEQ ID NO: 56

(LX5X6X7X8(W/F)X9XIOYXI 1X12G), wherein xs to X12 can be any naturally occurring amino acid residue, and xs particularly represents R or Q, xs particularly represents S, I or T, xn particularly represents E or S and X12 particularly represents C or M, in particular the motif of SEQ ID NOs: 57, 58, 59, 60 or 61, wherein said conserved sequence motif 1 corresponds to i. sequence positions 68 to 79 of SEQ ID NO: 3, ii. sequence positions 37 to 48 of SEQ ID NO: 6, iii. sequence positions 45 to 56 of SEQ ID NO: 9, iv. sequence positions 68 to 79 of SEQ ID NO: 12, v. sequence positions 28 to 39 of SEQ ID NO: 19, and vi. sequence positions 28 to 39 of SEQ ID NO: 26, respectively;

(2) the conserved sequence motif 2 set forth in SEQ ID NO: 62 (Yxi3Dxi4Xi5Rxi6RVD(P/A)V(V/A)xi7Xi8N), wherein XB to xis can be any naturally occurring amino acid residue, and X particularly represents F or L, and x½ particularly represents P or L, in particular the motif of SEQ ID NOs: 63, 64, 65, 66, 67 or 68, wherein said conserved sequence motif 2 corresponds to i. sequence positions 362 to 377 of SEQ ID NO: 3, ii. sequence positions 309 to 324 of SEQ ID NO: 6, iii. sequence positions 317 to 332 of SEQ ID NO: 9, iv. sequence positions 362 to 377 of SEQ ID NO: 12, v. sequence positions 299 to 314 of SEQ ID NO: 19, and vi. sequence positions 299 to 314 of SEQ ID NO: 26, respectively; and

(3) the conserved sequence motif 3 set forth in SEQ ID NO: 69 (GTxi9(Y/F)Yx20X2iX22Ex23FL(Y/F)), wherein XB to X23 can be any naturally occurring amino acid residue, and X19 particularly represents L or R, in particular the motif of SEQ ID NOs: 70, 71 or 72, wherein said conserved sequence motif 3 corresponds to i. sequence positions 410 to 422 of SEQ ID NO: 3, ii. sequence positions 357 to 369 of SEQ ID NO: 6, iii. sequence positions 365 to 377 of SEQ ID NO: 9, iv. sequence positions 410 to 422 of SEQ ID NO: 12, v. sequence positions 349 to 361 of SEQ ID NO: 19, and vi. sequence positions 349 to 361 of SEQ ID NO: 26, respectively.

5. The isolated polypeptide of anyone of the preceding claims, which a. comprises an amino acid sequence selected from SEQ ID NOs: 3, 6, 9, 12, 19, and

26; or b. is encoded by a nucleic acid molecule comprising a coding nucleotide sequence selected from SEQ ID NOs: 1, 2, 4, 5, 7, 8, 10, 11, 17, 18, 24 and 25.

6. An isolated nucleic acid molecule a. comprising a nucleotide sequence encoding the polypeptide of any one of the preceding claims; or b. comprising a nucleotide sequence selected from SEQ ID NOs: 1, 2, 4, 5, 7, 8, 10, 11, 17, 18, 24, and 25, or a nucleotide sequence having at least 40% sequence identity to the nucleotide sequence of SEQ ID NOs: 1, 2, 4, 5, 7, 8, 17, 18, 24, or 25 and encoding a polypeptide of the HAD-like hydrolase superfamily comprising terpene synthase activity; or c. comprising a nucleotide sequence comprising a sequence complementary to one of the sequences of b.; or d. comprising a nucleotide sequence hybridizing under stringent conditions to a nucleotide sequence of a., b. or c.

7. An expression construct comprising at least one nucleic acid molecule of claim 6.

8. A vector comprising at least one nucleic acid molecule of claim 6 or at least one expression construct of claim 7.

9. A recombinant non-human host cell or a recombinant non-human host organism comprising a. at least one isolated nucleic acid molecule of claim 6, optionally stably integrated into the genome; or b. at least one expression construct of claim 7, optionally stably integrated into the genome; or c. at least one vector of claim 8.

10. A method for producing at least one catalytically active polypeptide according to any one of claims 1 to 5 comprising: a. culturing a non-human host cell or non-human host organism of claim 9 to express at least one polypeptide according to anyone of claim 1 to 5; and b. optionally isolating the polypeptide from the non-human host cell or non-human organism cultured in step a.

11. A method for preparing a mutant polypeptide of the HAD-like hydrolase superfamily comprising terpene synthase activity, in particular drimenyl and/or albicanyl diphosphate synthase activity, which method comprises the steps of: a. selecting a nucleic acid molecule according to claim 6; b. modifying the selected nucleic acid molecule to obtain at least one mutant nucleic acid molecule; c. genetically modifying non-human host cells or unicellular non-human host organisms with the mutant nucleic acid sequence to express a polypeptide encoded by the mutant nucleic acid sequence, in particular by transformation; d. screening the expression product for at least one mutant comprising terpene synthase activity, in particular drimenyl and/or albicanyl diphosphate synthase activity and/or drimenol and/or albicanol synthase activity; and e. optionally, if the polypeptide has no desired mutant activity, repeat the process steps a. to d. until a polypeptide with a desired mutant activity is obtained; and f. optionally, if a polypeptide having a desired mutant activity was identified in step d., isolating the corresponding mutant nucleic acid obtained in step c.

12. A method for producing a drimane sesquiterpene, in particular drimenol and/or albicanol, comprising: a. contacting FPP with a polypeptide as defined in anyone of claims 1 to 5, or with a polypeptide prepared according to claim 10, or with a mutant polypeptide prepared according to claim 11, which polypeptide comprising terpene synthase activity, thereby obtaining at least one diphosphate of drimane sesquiterpene, in particular drimenyl diphosphate and/or albicanyl diphosphate; b. chemically or enzymatically cleaving the diphosphate moiety of said product obtained in step a.; and c. optionally isolating the drimane sesquiterpene, in particular drimenol and/or albicanol.

13. The method of claim 12, wherein the enzymatic step b. is performed by applying the same polypeptide as used in step a. which polypeptide comprises terpene synthase activity and phosphatase activity, or by applying a polypeptide different therefrom and having phosphatase enzyme.

14. The use of a polypeptide as defined in anyone of the claims 1 to 5 for preparing odorants, flavours or fragrance ingredients, in particular Ambrox.

15. A method of producing Ambrox, which method comprises providing drimenol and/or albicanol by a method of anyone of the claims as recited claim 12 or 13, optionally isolating drimenol and/or albicanol as produced in step a.; and converting drimenol and/or albicanol in a manner know per se to Ambrox.

Description:
NOVEL POLYPEPTIDES FOR PRODUCING ALBICANOL AND/OR DRIMENOL

COMPOUNDS

Technical Field

The present invention relates to novel plant-derived haloacid dehalogenase-like (HAD-like) polypeptides having cyclic terpene synthase (TPS) activity, in particular suitable for use in biochemical methods of producing drimane-type sesquiterpenes, encompassing drimenol and/or albicanol and related compounds, like phosphorylated drimane-type sesquiterpene alcohols, in particular phosphorylated drimenol and/or albicanol compounds and derivatives. The invention also provides the coding nucleotide sequences of said novel TPS activity, corresponding expression constructs, recombinant hosts, method of preparing such novel polypeptides and mutants and variants thereof. The invention also relates to the use of such novel polypeptides in the production of odorants, flavours and fragrance ingredients.

Background

Terpenes are found in most organisms (microorganisms, animals and plants). These compounds are made up of isoprene units and are classified by the number of these units present in their structure, which may comprise cyclic structural elements. Thus hemiterpenes, monoterpenes, sesquiterpenes and diterpenes are terpenes containing 5, 10, 15 and 20 carbon atoms (i. e. 1, 2, 3 and 4 isoprene units), respectively. Monoterpenes are derived from geranyl diphosphate (GPP, Cio), sesquiterpenes from famesyl diphosphate (FPP, C15), and diterpenes from geranylgeranyl diphosphate (GGPP, C20). Sesquiterpenes, for example, are widely found in the plant kingdom. Many sesquiterpene molecules are known for their flavour and fragrance properties and their cosmetic, medicinal and antimicrobial effects. Numerous sesquiterpene hydrocarbons and sesquiterpenoids have been identified. Chemical synthesis approaches have been developed but are still complex and not always cost-effective.

The biosynthetic production of terpenes involves enzymes called terpene synthases. There are numerous sesquiterpene synthases present in the plant kingdom, all using the same substrate (famesyl diphosphate, FPP), but having different product profiles. Genes and cDNAs encoding sesquiterpene synthases have been cloned and the corresponding recombinant enzymes characterized.

Many of the main sources for sesquiterpenes, for example, compounds with a drimane structure, like albicanol or drimenol compounds, are from plants; however, the content of sesquiterpenes in these natural sources can be low. There remains a need for the identification and characterization of new terpene synthases optionally characterized by a particular drimane-type product profile and/or drimane-type product yield and/or more cost-effective methods of producing drimane-type compounds, such as drimenol and/or albicanol, the potential building blocks for the preparation of highly valuable perfumery ingredients, such as Ambrox.

Summary

The above-mentioned problem could be solved by providing several new enzymes having cyclic terpene synthase (TPS) activity, in particular cyclic sesquiterpene synthase activity, and which more particularly show drimenyl and/or albicanyl diphosphate synthase activity and produce drimenyl and/or albicanyl diphosphate with surprisingly high selectivity from FPP. In particular, novel drimenyl and/or albicanyl diphosphate synthase genes were identified from plants of the genus Bazzania sp. such as Bazzania trilobata and from the plant Selaginella moellendorjfii. These new synthase enzymes are designated BazzHADl, BazzHAD2, BazzHAD3, BtHAD, SmHADl and SmHAD2.

The protein sequences of SmHADl and SmHAD2 have a very low identity with all other known HAD-like TPSs (-14-20%) but show high homology between themselves (>96%), BtHAD and BazzHADl show a slightly higher homology to fern and fungal HAD- like TPSs (-22-30%) and 96% identity between themselves, whereas BazzHAD2 and BazzHAD3 show an intermediate homology to fem and fungal HAD-like TPSs (-22-43%) and 67% identity between themselves (see Table 2 in experimental section).

All the HAD-like TPSs described herein contain a class II motif PxDxD(T/S)(T/M)S (SEQ ID NO:46), and more C-terminally shifted thereto a QW motif QxxDGx(W/F) (SEQ ID NO:51), mainly characteristics of diterpene synthases. Mutations performed on said class II motif cause full loss of the activity, mutation on said QW motif cause 90% loss of the activity. An class I motif (DDxx(D/E) (SEQ ID NO:45) is missing in all these six newly identified TPS of the present invention.

Description of the drawings.

Figure 1. a) Drimane structure; b) More generic drimane-type structure; the dotted line in the formula indicates that a single C=C double bond may be present in anyone the indicated positions Figure 2. Structural formula of (+)-albicanol and (-)-drimenol.

Figure 3. Mechanism of cyclization of famesyl diphosphate (FPP) by a HAD-like TPS containing class II motif and dephosphorylation of drimenyl and/or albicanyl diphosphate by the same HAD-like TPS or other phosphatase.

Figure 4. GC/MS chromatogram of samples from the in vivo biochemical assay of BazzHADl. Famesol results from the dephosphorylation of the precursor FPP, whereas drimenol is presumably the product of the dephosphorylation of drimenyl diphosphate.

Figure 5. Mass spectrum of the drimenol peak in Figure 4.

Figure 6. GC/MS chromatogram of samples from the in vivo biochemical assay of BazzHAD2. Famesol results from the dephosphorylation of the precursor FPP, whereas albicanol and unknown compound SQT1 are presumably the products of the dephosphorylation of albicanyl diphosphate and an unknown diphosphate, respectively.

Figure 7. Mass spectrum of the albicanol peak in Figure 4.

Figure 8. Mass spectrum of the unknown SQT1 peak in Figure 4.

Figure 9. GC/MS chromatogram of samples from the in vivo biochemical assay of BazzHAD3. Famesol results from the dephosphorylation of the precursor FPP, whereas albicanol is presumably the product of the dephosphorylation of albicanyl diphosphate.

Figure 10. Mass spectmm of the albicanol peak in Figure 7.

Figure 11. GC/MS chromatogram of samples from the in vivo biochemical assay of BtHAD. Famesol results from the dephosphorylation of the precursor FPP, whereas drimenol is presumably the product of the dephosphorylation of drimenyl diphosphate.

Figure 12. Mass spectmm of the drimenol peak in Figure 10.

Figure 13. GC/MS chromatogram of samples from the in vivo biochemical assay of SmHADl (EFJ10816.1). Famesol results from the dephosphorylation of the precursor FPP, whereas drimenol and albicanol are presumably the products of the dephosphorylation of drimenyl and albicanyl diphosphates, respectively.

Figure 14. Mass spectmm of the albicanol peak in Figure 11.

Figure 15. Mass spectmm of the drimenol peak in Figure 11. Figure 16. GC/MS chromatogram of samples from the in vivo biochemical assay of SmHAD2 (EFJ26126.1). Famesol results from the dephosphorylation of the precursor FPP, whereas drimenol and albicanol are presumably the products of the dephosphorylation of drimenyl and albicanyl diphosphates, respectively.

Figure 17. Mass spectrum of the albicanol peak in Figure 14.

Figure 18. Mass spectrum of the drimenol peak in Figure 14.

Figure 19. Comparison of yields (Mean ± SE, n=4~5) of drimane-type sesquiterpenes synthesized by SmHADl, SmHAD2, BtHAD, XP_007369631.1 and EMD37666.1 and their mutants in an in vivo biochemical assay. The SmHADl and SmHAD2 mutants comprise the mutations D257A and D259A in the class II motif (SEQ ID NOs: 21, 28 and 40) and the mutations E432A and D433A in the QW motif (SEQ ID NOs: 23, 30 and 44); the BtHAD mutant comprises the mutations D320A and D322A in the class II motif (SEQ ID NOs: 14 and 39) and the mutation D491A in the QW motif (SEQ ID NOs: 16 and 43); the EMD37666.1 mutant comprises the mutations D276A, D277A, and D279A in the class II motif (SEQ ID NOs: 34 and 41); the XP_007369631.1 mutant comprises the mutations D272A, D273A and D275A in the class II motif (SEQ ID NOs: 38 and 42).

Figure 20. GC/MS chromatograms of samples from the in vitro biochemical assay of SmHADl, SmHAD2 and BtHAD in the absence or presence of BAP (Bacterial alkaline phosphatase). (2 26E)-Farncsol results from the dephosphorylation of the precursor FPP, whereas albicanol and drimenol are presumably the products of the dephosphorylation of albicanyl and drimenyl diphosphates, respectively.

Figure 21. Amino acid sequences alignment of HAD-like TPSs: BazzHADl (SEQ ID NO: 3), BazzHAD2 (SEQ ID NO: 6), BazzHAD3 (SEQ ID NO: 9), BtHAD (SEQ ID NO: 12), SmHADl (SEQ ID NO: 19), SmHAD2 (SEQ ID NO: 26); EMD37666.1 (SEQ ID NO: 32) and XP_007369631.1 (SEQ ID NO: 36) as previously described in WO 2018/220113. The class I motif (DDxx(D/E); SEQ ID NO: 45) and class II motif (PxDxD(T/S)(T/M)S; SEQ ID NOs: 46) as well as three other conserved sequence motifs 1 - 3 (Lxxxx(W/F)xxYxxG; SEQ ID NO: 56, YxDxxRxRVD(P/A)V(V/A)xxN; SEQ ID NO: 62, GTx(Y/F)YxxxExFL(Y/F); SEQ ID NO: 69) are identified in the Figure.

Abbreviations used bp base pair BAP bacterial alkaline phosphatase

BSA bovine serum albumin

DNA deoxyribonucleic acid cDNA complementary DNA

DTT dithiothreitol

FPP famesyl diphosphate

GC gas chromatograph

HAD haloacid dehalogenase

IPTG isopropyl-D-thiogalacto-pyranoside

LB lysogeny broth

MS mass spectrometer / mass spectrometry

MVA mevalonic acid

PCR polymerase chain reaction

PP diphosphate or pyrophosphate

RNA ribonucleic acid mRNA messenger ribonucleic acid miRNA micro RNA siRNA small interfering RNA rRNA ribosomal RNA tRNA transfer RNA sp. Species

TPS Terpene Synthase

Detailed Description a. Definitions

“Terpenes” are a large and diverse class of organic compounds, produced by a variety of plants, particularly conifers, and by some insects. Terpenes are hydrocarbons. Although sometimes used interchangeably with "terpenes", “terpenoids” or “isoprenoids” are modified terpenes as they contain additional functional groups, usually oxygen-containing.

“Terpenoids” (“isoprenoids”) are a large and diverse class of naturally occurring organic chemicals derived from terpenes. Although sometimes used interchangeably with the term “terpenes”, “terpenoids” contain additional functional groups, usually O-containing groups, like for example hydroxyl, carbonyl or carboxyl groups. Most are multicyclic structures with oxygen-containing functional groups. Unless stated otherwise, in the context of the present description the term “terpene” and the term “terpenoid” may be used interchangeably.

Terpenes (and terpenoids) may be classified by the number of isoprene units in the molecule; a prefix in the name indicates the number of terpene units needed to assemble the molecule. Hemiterpenes consist of a single isoprene unit. Monoterpenes consist of two isoprene units and have the molecular formula C10H16. Sesquiterpenes consist of three isoprene units and have the molecular formula C15H24. Diterpenes are composed of four isoprene units and have the molecular formula C20H32.

“Drimane” or “drimane sesquiterpene” is a bicyclic sesquiterpene and is the parent structure of many natural products with various biological activities (Figure la). According to IUPAC its systematic chemical name is (4aR,5S,6S,8aS)-l,l,4a,5,6- pentamethyldecahydronaphthalene. For purposes of this application the term “drimane” has to be understood more broadly, and is also referred to as “drimane-type”, and covers bicyclic sesquiterpenes having the general structures as depicted in Figure lb. It is not limited to a particular stereochemistry and may contain a single C=C-double bond at certain positions of the drimane skeleton. Particular drimane structures are those as contained in drimenol and albicanol.

“Drimane alcohol” or “drimane sesquiterpene alcohol” refer to a “Drimane” or “drimane sesquiterpene” carrying a hydroxyl group; particular examples are drimenol and albicanol. The term relates in particular to a cyclic terpene (terpenoid) having a drimane-like carbon skeleton structure as depicted in Figure lb.

“Albicanol” for the purpose of this application relates to (-i-)-albicanol (CAS: 54632- 04-1) (Figure 2).

“Albicanol derivatives” include, for example, compounds derived from albicanol by one or more steps of esterification, hydroxylation, oxidation, acylation, isomerization, dimethylation and the like. Suitable derivatives may be selected from a hydrocarbon, alcohol, diol, triol, acetal, ketal, aldehyde, acid, ether, amide, ketone, lactone, epoxide, acetate, glycoside and/or an ester.

“Drimenol” for purposes of this application relates to (-)-drimenol (CAS: 468-68-8) (Figure 2).

“Drimenol derivatives” include, for example, compounds derived from drimenol by or more steps of esterification, hydroxylation, oxidation, acylation, isomerization, dimethylation and the likes. Suitable derivatives may be selected from a hydrocarbon, alcohol, diol, triol, acetal, ketal, aldehyde, acid, ether, amide, ketone, lactone, epoxide, acetate, glycoside and/or an ester.

“Ambrox” for purposes of this application relates to IUPAC Name: (-)- (3aR,5aS,9aS,9bR)-3a,6,6,9a-tetramethyldodecahydronaphto[2,l -b]furan (CAS : 6790-58-5).

A “precursor” molecule of a target compound as described herein is converted to said target compound, preferably through the enzymatic action of a suitable polypeptide performing at least one structural change on said precursor molecule. For example a “diphosphate precursor” (as for example a “terpenyl diphosphate precursor”) is converted to said target compound (as for example a terpene alcohol) via enzymatic removal of the diphosphate moiety, for example by removal of mono- or diphosphate groups by a phosphatase enzyme. For example a “non-cyclic precursor” (like a “non-cyclic terpenyl precursor”) may be converted to the cyclic target molecule (like a cyclic terpene compound) through the action of a cyclase or synthase enzyme, irrespective of the particular enzymatic mechanism of such enzyme, in one or more steps.

The terms “cyclic terpene synthase” or “polypeptide having cyclic terpene synthase activity” are used as synonyms to the terms “terpene cyclase” or “polypeptide having terpene cyclase activity”. The term “TPS” is used as abbreviation to the term “terpene synthase”. A cyclic terpene synthase as described herein belongs to the haloacid dehalogenase-like (HAD- like) hydrolase superfamily which comprises domains corresponding to Pfam domains PF13419 (http://pfam.xfam.org/family/PF13419) and/or PF00702

(http://pfam.xfam.org/family/PF00702.26). Some cyclic terpene synthases herein which don’t have a significant Pfam domains PF13419 and/or PF00702 remain considered as from HAD- like hydrolase as harboring modified conserved motifs of the above mentioned HAD-like hydrolase superfamily, e.g. class I motif location, class II motif location and QW motif location (SEQ ID: 45, SEQ ID: 46, SEQ ID: 51). A HAD-like hydrolase domain is a portion of a polypeptide having amino acid sequence similarities with the members of the HAD-like hydrolase family and related function. A HAD-like hydrolase domain can be identified in a polypeptide by searching for amino acid motifs or signatures characteristic of this protein family. Tools for performing such searches are available, for example, at the following web sites: https://www.ebi.ac.uk/interpro/ or https://www.ebi.ac.uk/Tools/hmmer/. Proteins are generally composed of one or more functional regions or domains. Different combinations of domains give rise to the diverse range of proteins found in nature. The identification of domains that occur within proteins can therefore provide insights into their function. A polypeptide which comprises a HAD-like hydrolase domain and/or characteristic HAD-like hydrolase motifs functions in binding and cleavage of phosphate or diphosphate groups of a ligand. An polypeptide of the haloacid dehalogenase-like (HAD-like) hydrolase superfamily, comprising cyclic terpene synthase activity could be defined as HAD-like TPS.

The term “class I terpene synthase” relates to a terpene synthase that catalyzes ionization-initiated reactions, for example, monoterpene and sesquiterpene synthases.

The term “class I terpene synthase motif’, “class I terpene synthase-like motif’, “class I synthase(-like) motif’, “class I synthase motif’ or “class I motif’ relates to an active site of a terpene synthase that comprises the conserved DDxx(D/E) motif (SEQ ID NO: 45). The aspartic acid residues of this class I motif bind, for example, a divalent metal ion (most often Mg 2 + ) involved in the binding of the diphosphate group and catalyze the ionization and cleavage of the allylic diphosphate bond of the substrate.

The term “class II terpene synthase” or “class II synthase” relates to a terpene synthase that catalyzes protonation-initiated cyclization reaction, for example, typically involved in the biosynthesis of triterpenes and labdane diterpenes. In class II terpene synthases, the protonation-initiated reaction may involve, for example, acidic amino acids donating a proton to the terminal double-bond.

The term “(modified) class II terpene synthase motif’, “ (modified) class II terpene synthase-like motif’, “(modified) class II synthase(-like) motif’, “(modified) class II synthase motif’ or “(modified) class II motif’ relates to an active site of a terpene synthase that comprises the conserved DxDxxS (SEQ ID NO: 75) motif, e.g. PxDxD(T/S)(T/M)S motif (SEQ ID NO: 46) herein.

The term “QW motif’ herein relates to an active site of a terpene synthase that comprises the conserved QxxxxxW (SEQ ID NO: 76) motif, e.g. QxxDGxW motif (SEQ ID NO: 50) herein.

The terms “albicanyl diphosphate synthase” or “polypeptide having albicanyl diphosphate synthase activity” or “albicanyl diphosphate synthase protein” or “having the ability to produce albicanyl diphosphate” relate to a polypeptide capable of catalyzing the synthesis of albicanyl diphosphate, in the form of any of its stereoisomers or a mixture thereof, starting from an acyclic terpene pyrophosphate, particularly famesyl diphosphate (FPP). Albicanyl diphosphate may be the only product or may be part of a mixture of sesquiterpenes. Said mixture may comprise albicanyl monophosphate and/or albicanol.

The terms “albicanol synthase” or “polypeptide having a albicanol synthase activity” or “albicanol synthase protein” relate to a polypeptide capable of catalyzing the synthesis of albicanol, in the form of any of its stereoisomers or a mixture thereof, starting from an acyclic terpene pyrophosphate, particularly famesyl diphosphate (FPP). Albicanol may be the only product or may be part of a mixture of sesquiterpenes.

“Albicanyl diphosphate synthase activity” is determined under “standard conditions” as described herein below: They can be determined using recombinant albicanyl diphosphate synthase expressing host cells, disrupted albicanyl diphosphate synthase expressing cells, fractions of these or enriched or purified albicanyl diphosphate synthase enzyme, in a culture medium or reaction medium, preferably buffered, having a pH in the range of 6 to 11, preferably 7 to 9, at a temperature in the range of about 20 to 45°C, like about 25 to 40°C, preferably 25 to 32°C and in the presence of a reference substrate, here in particular FPP, either added at an initial concentration in the range of 1 to 100 mM, preferably 5 to 50 mM, in particular 30 to 40 pM, or endogenously produced by the host cell. The conversion reaction to form of albicanyl diphosphate is conducted form 10 min to 5 h, preferably about 1 to 2 h. If no endogenous alkaline phosphatase is present, one or more exogenous phosphatase is added to the reaction mixture to convert albicanyl diphosphate as formed by the synthase. Albicanol may then be determined in conventional manner, for example after extraction with an organic solvent, like ethyl acetate. Particular examples of suitable standard conditions are applied in the Experimental Part below, like in Example 4 below, which conditions also shall form part of the general disclosure of the invention.

The terms “drimenyl diphosphate synthase” or “polypeptide having drimenyl diphosphate synthase activity” or “drimenyl diphosphate synthase protein” or “having the ability to produce drimenyl diphosphate” relate to a polypeptide capable of catalyzing the synthesis of drimenyl diphosphate, in the form of any of its stereoisomers or a mixture thereof, starting from an acyclic terpene pyrophosphate, particularly famesyl diphosphate (FPP). Drimenyl diphosphate may be the only product or may be part of a mixture of sesquiterpenes. Said mixture may comprise drimenyl monophosphate and/or drimenol.

The terms “drimenol synthase” or “polypeptide having a drimenol synthase activity” or “drimenol synthase protein” relate to a polypeptide capable of catalyzing the synthesis of drimenol, in the form of any of its stereoisomers or a mixture thereof, starting from an acyclic terpene pyrophosphate, particularly famesyl diphosphate (FPP). Drimenol may be the only product or may be part of a mixture of sesquiterpenes.

“Drimenyl diphosphate synthase activity” is determined under “standard conditions” as described herein below: They can be determined using recombinant drimenyl diphosphate synthase expressing host cells, dismpted drimenyl diphosphate synthase expressing cells, fractions of these or enriched or purified drimenyl diphosphate synthase enzyme, in a culture medium or reaction medium, preferably buffered, having a pH in the range of 6 to 11, preferably 7 to 9, at a temperature in the range of about 20 to 45°C, like about 25 to 40°C, preferably 25 to 32°C and in the presence of a reference substrate, here in particular FPP, either added at an initial concentration in the range of 1 to 100 mM, preferably 5 to 50 mM, in particular 30 to 40 pM, or endogenously produced by the host cell. The conversion reaction to form of drimenyl diphosphate is conducted form 10 min to 5 h, preferably about 1 to 2 h. If no endogenous alkaline phosphatase is present, one or more exogenous phosphatase is added to the reaction mixture to convert drimenyl diphosphate as formed by the synthase. Drimenol may then be determined in conventional manner, for example after extraction with an organic solvent, like ethyl acetate. Particular examples of suitable standard conditions are applied in the Experimental Part below, like in Example 4 below, which conditions also shall form part of the general disclosure of the invention.

The terms “biological function”, “function”, “biological activity”, or “activity” refer to the ability of the terpene synthases as described herein to catalyze the formation of drimenyl diphosphate and/or drimenol, and/or albicanyl diphosphate and/or albicanol; or a mixture of compounds comprising drimenyl diphosphate and/or drimenyl monophosphate and/or drimenol, and/or albicanyl diphosphate and/or albicanyl monophosphate and/or albicanol, and/or and one or more other terpenes, in particular drimenyl diphosphate and/or albicanyl diphosphate.

The terms “mixture of terpenes” or “mixture of sesquiterpenes” refer to a mixture of terpenes or of sesquiterpenes that comprises drimenyl diphosphate and/or drimenyl monophosphate and/or drimenol, and/or albicanyl diphosphate and/or albicanyl monophosphate and/or albicanol and may also comprise one or more additional terpenes, like one or more additional sesquiterpenes.

The “mevalonate pathway” also known as the “isoprenoid pathway” or “HMG-CoA reductase pathway” is an essential metabolic pathway present in eukaryotes, archaea, and some bacteria. The mevalonate pathway begins with acetyl-CoA and produces two five- carbon building blocks called isopentenyl pyrophosphate (IPP) and dimethylallyl pyrophosphate (DMAPP). Key enzymes are acetoacetyl-CoA thiolase (atoB), HMG-CoA synthase (mvaS), HMG-CoA reductase (mvaA), mevalonate kinase (MvaKl), phosphomevalonate kinase (MvaK2), a mevalonate diphosphate decarboxylase (MvaD), and an isopentenyl diphosphate isomerase (idi). Combining the mevalonate pathway with enzyme activity to generate the terpene precursors GPP, FPP or GGPP, like in particular FPP synthase (ERG20), allows the recombinant cellular production of terpenes. As used herein, the term “recombinant host cell/organism”, “genetically altered (modified) cell/organism” or “transformed cell/organism” refers to a cell or organism altered to harbor at least one nucleic acid molecule, for instance, a recombinant gene encoding a desired protein or nucleic acid sequence which upon transcription yields a functional polypeptide of the present invention, in particular a drimenyl diphosphate synthase protein useful to produce drimenyl diphosphate and/or drimenyl monophosphate and/or drimenol or corresponding mixtures of terpenes containing drimenyl diphosphate and/or drimenyl monophosphate and/or drimenol, and/or an albicanyl diphosphate synthase protein useful to produce albicanyl diphosphate and/or albicanyl monophosphate and/or albicanol or corresponding mixtures of terpenes containing albicanyl diphosphate and/or albicanyl monophosphate and/or albicanol. The host cell is particularly a bacterial cell, a fungal cell, a yeast cell or a plant cell, or plants. The host cell may contain a recombinant gene which has been integrated into the nuclear or organelle genomes of the host cell. Alternatively, the host may contain the recombinant gene extra- chromosomally.

The term “organism” refers to any non-human multicellular or unicellular organisms such as a plant, or a microorganism. Particularly, a microorganism is a bacterium, a yeast, an algae or a fungus.

The term “plant” is used interchangeably to include plant cells including plant protoplasts, plant tissues, plant cell tissue cultures giving rise to regenerated plants, or parts of plants, or plant organs such as roots, stems, leaves, flowers, pollen, ovules, embryos, fruits and the like. Any plant can be used to carry out the methods of an embodiment herein.

A particular organism or cell is meant to be “capable of producing FPP” when it produces FPP naturally or when it does not produce FPP naturally but is (genetically) modified, such as transformed, to produce FPP with a nucleic acid as described herein. Organisms or cells transformed to produce a higher or lower, in particular higher amount of FPP than the naturally occurring organism or cell are also encompassed by the “organisms or cells capable of producing FPP”.

A particular organism or cell is meant to be “capable of producing albicanyl diphosphate” when it produces albicanyl diphosphate naturally or when it does not produce albicanyl diphosphate naturally but is transformed to produce albicanyl diphosphate with a nucleic acid as described herein. Organisms or cells transformed to produce a higher or lower, in particular higher amount of albicanyl diphosphate than the naturally occurring organism or cell are also encompassed by the “organisms or cells capable of producing albicanyl diphosphate”. A particular organism or cell is meant to be “capable of producing albicanol” when it produces albicanol naturally or when it does not produce albicanol naturally but is transformed to produce albicanyl diphosphate with a nucleic acid as described herein, and optionally further transformed with a nucleic acid to produce enzyme activity converting albicanyl diphosphate to albicanol. Organisms or cells transformed to produce a higher or lower, in particular higher amount of albicanol than the naturally occurring organism or cell are also encompassed by the “organisms or cells capable of producing albicanol”.

A particular organism or cell is meant to be “capable of producing drimenyl diphosphate” when it produces drimenyl diphosphate naturally or when it does not produce drimenyl diphosphate naturally but is transformed to produce drimenyl diphosphate with a nucleic acid as described herein. Organisms or cells transformed to produce a higher or lower, in particular higher amount of drimenyl diphosphate than the naturally occurring organism or cell are also encompassed by the “organisms or cells capable of producing drimenyl diphosphate”.

A particular organism or cell is meant to be “capable of producing drimenol” when it produces drimenol naturally or when it does not produce drimenol naturally but is transformed to produce drimenol diphosphate with a nucleic acid as described herein, and optionally further transformed with a nucleic acid to produce enzyme activity converting drimenyl diphosphate to drimenol. Organisms or cells transformed to produce a higher or lower, in particular higher amount of drimenol than the naturally occurring organism or cell are also encompassed by the “organisms or cells capable of producing drimenol”.

The terms “purified", “substantially purified”, and “isolated” as used herein refer to the state of being free of other, dissimilar compounds with which a compound of the invention is normally associated in its natural state, so that the “purified”, “substantially purified”, and “isolated” subject comprises at least 0.5%, 1%, 5%, 10%, or 20%, or at least 50% or 75% of the mass, by weight, of a given sample. In one embodiment, these terms refer to the compound of the invention comprising at least 95, 96, 97, 98, 99 or 100%, of the mass, by weight, of a given sample. As used herein, the terms “purified", “substantially purified”, and “isolated” when referring to a nucleic acid or protein, of nucleic acids or proteins, also refers to a state of purification or concentration different than that which occurs naturally, for example in an prokaryotic or eukaryotic environment, like, for example in a bacterial or fungal cell, or in the mammalian organism, especially human body. Any degree of purification or concentration greater than that which occurs naturally, including (i) the purification from other associated structures or compounds or (ii) the association with structures or compounds to which it is not normally associated in said prokaryotic or eukaryotic environment, are within the meaning of "isolated”. The nucleic acid or protein or classes of nucleic acids or proteins, described herein, may be isolated, or otherwise associated with structures or compounds to which they are not normally associated in nature, according to a variety of methods and processes known to those of skill in the art.

The term “about” indicates a potential variation of ± 25% of the stated value, in particular ± 15%, ± 10 %, more particularly ± 5%, ± 2% or ± 1%.

The term “substantially” describes a range of values of from about 80 to 100%, such as, for example, 85-99.9%, in particular 90 to 99.9%, more particularly 95 to 99.9%, or 98 to 99.9% and especially 99 to 99.9%.

“Predominantly” refers to a proportion in the range of above 50%, as for example in the range of 51 to 100%, particularly in the range of 75 to 99,9%; more particularly 85 to 98,5%, like 95 to 99%.

A “main product” in the context of the present invention designates a single compound or a group of at least 2 compounds, like 2, 3, 4, 5 or more, particularly 2 or 3 compounds, which single compound or group of compounds is “predominantly” prepared by a reaction as described herein, and is contained in said reaction in a predominant proportion based on the total amount of the constituents of the product formed by said reaction. Said proportion may be a molar proportion, a weight proportion or, preferably based on chromatographic analytics, an area proportion calculated from the corresponding chromatogram of the reaction products.

A “side product” in the context of the present invention designates a single compound or a group of at least 2 compounds, like 2, 3, 4, 5 or more, particularly 2 or 3 compounds, which single compound or group of compounds is not “predominantly” prepared by a reaction as described herein.

Because of the reversibility of enzymatic reactions, the present invention relates, unless otherwise stated, to the enzymatic or biocatalytic reactions described herein in both directions of reaction.

“Functional mutants” of herein described polypeptides include the “functional equivalents” of such polypeptides as defined below.

The term “stereoisomers” includes in particular conformational isomers.

Included in general are, according to the invention, all “stereoisomeric forms” of the compounds described herein, such as “constitutional isomers” and, in particular, “stereoisomers”. “Stereoisomeric forms” encompass in particular, “stereoisomers” and mixtures thereof, e.g. configuration isomers (optical isomers), such as enantiomers, or geometric isomers (diastereomers), such as E- and Z-isomers, and combinations thereof. If one or more asymmetric centers are present in one molecule, the invention encompasses all combinations of different conformations of these asymmetry centers, e.g. enantiomeric pairs.

“Stereoselectivity” describes the ability to produce a particular stereoisomer of a compound in a stereoisomerically pure form or to specifically convert a particular stereoisomer in an enzyme catalyzed method as described herein out of a plurality of stereoisomers. More specifically, this means that a product of the invention is enriched with respect to a specific stereoisomer, or an educt may be depleted with respect to a particular stereoisomer. This may be quantified via the purity %ee-parameter calculated according to the formula:

%ee = [XA-XB]/[ XA+XB]*100, wherein XA and XB represent the molar ratio (Molenbmch) of the stereoisomers A and B.

The terms “selectively converting” or “increasing the selectivity” in general means that a particular stereoisomeric form, as for example the E-form, of an unsaturated hydrocarbon, is converted in a higher proportion or amount (compared on a molar basis) than the corresponding other stereoisomeric form, as for example Z-form, either during the entire course of said reaction (i.e. between initiation and termination of the reaction), at a certain point of time of said reaction, or during an “interval” of said reaction. In particular, said selectivity may be observed during an “interval” corresponding to 1 to 99%, 2 to 95%, 3 to 90%, 5 to 85%, 10 to 80%, 15 to 75%, 20 to 70%, 25 to 65%, 30 to 60%, or 40 to 50% conversion of the initial amount of the substrate. Said higher proportion or amount may, for example, be expressed in terms of: a higher maximum yield of an isomer observed during the entire course of the reaction or said interval thereof; a higher relative amount of an isomer at a defined % degree of conversion value of the substrate; and/or an identical relative amount of an isomer at a higher % degree of conversion value; each of which preferably being observed relative to a reference method, said reference method being performed under otherwise identical conditions with known chemical or biochemical means. Generally also comprised in accordance with the invention are all “isomeric forms” of the compounds described herein, such as constitutional isomers and in particular stereoisomers and mixtures of these, such as, for example, optical isomers or geometric isomers, such as E- and Z-isomers, and combinations of these. If several centers of asymmetry are present in a molecule, then the invention comprises all combinations of different conformations of these centers of asymmetry, such as, for example, pairs of enantiomers, or any mixtures of stereoisomeric forms.

For the descriptions herein and the appended claims, the use of “or” means “and/or” unless stated otherwise. Similarly, “comprise”, “comprises”, “comprising”, “include”, “includes”, and “including” are interchangeable and not intended to be limiting.

It is to be further understood that where descriptions of various embodiments use the term "comprising", those skilled in the art would understand that in some specific instances an embodiment can be alternatively described using language "consisting essentially of" or "consisting of’.

If the present disclosure refers to features, parameters and ranges thereof of different degree of preference (including general, not explicitly preferred features, parameters and ranges thereof) then, unless otherwise stated, any combination of two or more of such features, parameters and ranges thereof, irrespective of their respective degree of preference, is encompassed by the disclosure of the present description. b. Particular embodiments of the invention

The present invention relates to the following embodiments:

1. An isolated polypeptide of the haloacid dehalogenase-like (HAD-like) hydrolase superfamily, comprising cyclic terpene synthase activity, wherein said polypeptide is selected from a. BazzHADl comprising an amino acid sequence of SEQ ID NO: 3, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 3 and retaining said cyclic terpene synthase activity, in particular drimenyl diphosphate synthase activity; b. BazzHAD2 comprising an amino acid sequence of SEQ ID NO: 6, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 6 and retaining said cyclic terpene synthase activity, in particular albicanyl diphosphate synthase activity; c. BazzHAD3 comprising an amino acid sequence of SEQ ID NO: 9, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 9 and retaining said cyclic terpene synthase activity, in particular albicanyl diphosphate synthase activity; d. BtHAD comprising an amino acid sequence of SEQ ID NO: 12, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 12 and retaining said cyclic terpene synthase activity, in particular drimenyl diphosphate synthase activity; e. SmHADl comprising an amino acid sequence of SEQ ID NO: 19, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 19 and retaining said cyclic terpene synthase activity, in particular drimenyl diphosphate synthase activity and/or in particular albicanyl diphosphate synthase activity ; f. SmHAD2 comprising an amino acid sequence of SEQ ID NO: 26, or a mutant or natural variant thereof comprising an amino acid sequence having at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 26 and retaining said cyclic terpene synthase activity, in particular drimenyl diphosphate synthase activity and/or in particular albicanyl diphosphate synthase activity;

A particular subgroup of TPSs refers to TPSs listed above under a), b), c) and d), more particularly a), b) and c).

A more particular aspect of this embodiment relates to polypeptide variants or in particular non-natural mutants of the above novel polypeptides of the invention which comprise cyclic terpene synthase activity as identified above. These variants or non-natural mutants are derived from anyone of the particular amino acid sequences of SEQ ID NOs: 3, 6, 9, 12, 19, and 26. These variants are selected from polypeptides comprising an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to anyone of SEQ ID NO: 3, 6, 9, 12, 19, and 26, which contain at least one amino acid sequence difference, such as at least one amino acid sequence position which is modified by addition, substitution, insertion or deletion relative to the respective non- modified polypeptide of SEQ ID NOs: 3, 6, 9, 12, 19 or 26. Thus such variants have less than 100% sequence identity to the respective non-modified polypeptide. “At least one” in this context encompasses at least 1 to 20, 1 to 15, 1 tolO, and more particularly 1, 2, 3, 4 or 5 amino acid sequence positions independently of each other modified by amino acid residue addition, substitution, insertion or deletion.

2. The polypeptide of embodiment 1, comprising the ability to produce a) a phosphate precursor of at least one drimane sesquiterpene alcohol; or b) the phosphate precursor of at least one drimane sesquiterpene alcohol and the corresponding at least one drimane sesquiterpene alcohol; or c) at least one drimane sesquiterpene alcohol.

3. The polypeptide of embodiment 2, wherein the phosphate precursor is a drimanyl monophosphate, or, more particularly, a drimanyl diphosphate; and/or wherein the drimane sesquiterpene alcohol is drimenol and/or albicanol.

4. The polypeptide of embodiment 1, 2 or 3, comprising the ability to produce a) drimenyl diphosphate and/or albicanyl diphosphate from famesyl diphosphate (FPP) as substrate; or b) drimenyl diphosphate and/or albicanyl diphosphate from famesyl diphosphate (FPP) as substrate; and drimenol and/or albicanol, either directly from FPP as substrate or via the respective diphosphate precursor; or c) drimenol and/or albicanol from FPP as substrate, either directly from FPP as substrate or via the respective diphosphate precursor.

In the following Table 1 an overview of particular TPS of the invention, their particular sequence motifs including their relative position in the full amino acid sequence and their respective product profile (based on type of drimane alcohol) is shown.

5. The polypeptide of one of the preceding embodiments, which is isolated from a. plants of the genus Bazzania, in particular of the species Bazzania trilobata or b. plants of the genus Selaginella, in particular of the species Selaginella moellendorjfii c. other lycophytes, liverworts or monilophytes that contain drimane sesquiterpenes, including but not limited to Porella sp., Hymenophyton sp., and Marchantia sp.; or from other plant species.

6. The polypeptides of anyone of embodiments 1 to 5 further comprising a. a modified class II synthase motif as set forth in SEQ ID NO: 46 (PxoDxiD(T/S)(T/M)S), wherein xo and xi can be any naturally occurring amino acid residue, and xi particularly represents I or L, in particular the motif of any one of SEQ ID NOs: 47, 48, 49 or 50, wherein said modified class II synthase motif corresponds to i. sequence positions 318 to 325 of SEQ ID NO: 3, ii. sequence positions 269 to 276 of SEQ ID NO: 6, iii. sequence positions 277 to 284 of SEQ ID NO: 9, iv. sequence positions 318 to 325 of SEQ ID NO: 12, v. sequence positions 255 to 262 of SEQ ID NO: 19, and vi. sequence positions 255 to 262 of SEQ ID NO: 26, respectively; b. a QW motif as set forth in SEQ ID NO: 51 (QX2X3DGX4W), wherein X2, X3 and X4 can be any naturally occurring amino acid residue, and X4 particularly represents G or S, in particular the motif of any one of SEQ ID NOs: 52, 53, 54, or 55, wherein said QW motif corresponds to i. sequence positions 489 to 495 of SEQ ID NO: 3, ii. sequence positions 431 to 437 of SEQ ID NO: 6, iii. sequence positions 439 to 445 of SEQ ID NO: 9, iv. sequence positions 488 to 494 of SEQ ID NO: 12, v. sequence positions 430 to 436 of SEQ ID NO: 19, and vi. sequence positions 430 to 436 of SEQ ID NO: 26, respectively; and c. optionally at least one, as for example 1, 2 or 3, further sequence motif selected from (1) the conserved sequence motif 1 set forth in SEQ ID NO: 56

(Lx5X6X7X8(W/F)x9XioYxiixi2G), wherein xs to X12 can be any naturally occurring amino acid residue, and xs particularly represents R or Q, xs particularly represents S, I or T, xn particularly represents E or S and X12 particularly represents C or M, in particular the motif of SEQ ID NOs: 57, 58, 59, 60 or 61, wherein said conserved sequence motif 1 corresponds to i. sequence positions 68 to 79 of SEQ ID NO: 3, ii. sequence positions 37 to 48 of SEQ ID NO: 6, iii. sequence positions 45 to 56 of SEQ ID NO: 9, iv. sequence positions 68 to 79 of SEQ ID NO: 12, v. sequence positions 28 to 39 of SEQ ID NO: 19, and vi. sequence positions 28 to 39 of SEQ ID NO: 26, respectively;

(2) the conserved sequence motif 2 set forth in SEQ ID NO: 62

(Yxi3Dxi4Xi5Rxi6RVD(P/A)V(V/A)xi7XisN), wherein xu to xis can be any naturally occurring amino acid residue, and X B particularly represents F or L, and xi 6 particularly represents P or L, in particular the motif of SEQ ID NOs: 63, 64, 65, 66, 67 or 68, wherein said conserved sequence motif 2 corresponds to i. sequence positions 362 to 377 of SEQ ID NO: 3, ii. sequence positions 309 to 324 of SEQ ID NO: 6, iii. sequence positions 317 to 332 of SEQ ID NO: 9, iv. sequence positions 362 to 377 of SEQ ID NO: 12, v. sequence positions 299 to 314 of SEQ ID NO: 19, and vi. sequence positions 299 to 314 of SEQ ID NO: 26, respectively; and

(3) the conserved sequence motif 3 set forth in SEQ ID NO: 69

(GTxi9(Y/F)Yx20X2iX22Ex23FL(Y/F)), wherein X19 to X23 can be any naturally occurring amino acid residue, and X19 particularly represents L or R, in particular the motif of SEQ ID NOs: 70, 71 or 72, wherein said conserved sequence motif 3 corresponds to i. sequence positions 410 to 422 of SEQ ID NO: 3, ii. sequence positions 357 to 369 of SEQ ID NO: 6, iii. sequence positions 365 to 377 of SEQ ID NO: 9, iv. sequence positions 410 to 422 of SEQ ID NO: 12, v. sequence positions 349 to 361 of SEQ ID NO: 19, and vi. sequence positions 349 to 361 of SEQ ID NO: 26, respectively.

More particularly, a TPS of the present invention comprises a class II motif, a QW motif, and at least one of the conserved motifs 1, 2 and 3. Even more particularly, a TPS of the present invention comprises a class II motif, a QW motif, and at least two of the conserved motifs 1, 2 and 3.

Still more particularly, a TPS of the present invention comprises a class II motif, a QW motif, and all three of the conserved motifs 1, 2 and 3.

The relative order of these sequence motifs of embodiment 6 within the respective amino acid sequences of each of the TPSs of the present invention is depicted below (see also Figure 21):

N-terminus — (conserved motif 1) — (class II motif) — (conserved motif 2) — —(conserved motif 3) — (QW motif) — C-terminus

The class I motif known from other TPS, which is missing in TPS of the invention would be located between Conserved Motif 1 and class II motif (see Figure 21).

7. The polypeptide of anyone of the preceding embodiments, which catalyzes the conversion of an acyclic farnesyl diphosphate (in particular of (2F,6F)-3,7,1 1- trimethyldodeca-2,6,10-triene-l-pyrophosphate; FPP) to an drimenyl phosphate derivative like a monophosphate and more particularly an drimenyl diphosphate, in particular with a selectivity of 50%ee or more, like 50 to 100%ee, or 60 to 90%ee or 70 to 80 %ee.

8. The polypeptide of anyone of the preceding embodiments, which catalyses the conversion of an acyclic farnesyl diphosphate (in particular of (2£ , ,6£)-3,7,ll- trimethyldodeca-2,6,10-triene-l-pyrophosphate; FPP) to an albicanyl phosphate derivative like a monophosphate and more particularly an albicanyl diphosphate, in particular with a selectivity of 50%ee or more, like 50 to 100%ee, or 60 to 90%ee or 70 to 80 %ee..

9. The isolated polypeptide of anyone of the preceding embodiments, which a. comprises an amino acid sequence selected from SEQ ID NOs: 3, 6, 9, 12, 19, and 26; or b. is encoded by a nucleic acid molecule comprising a coding nucleotide sequence selected from SEQ ID NOs: 1, 2, 4, 5, 7, 8, 10, 11, 17, 18, 24 and 25.

10. The isolated polypeptide of embodiment 9, which a. consists of an amino acid sequence selected from SEQ ID NOs: 3, 6, 9, 12, 19, and 26; or b. is encoded by a nucleic acid molecule consisting of a coding nucleotide sequence selected from SEQ ID NOs: 1, 2, 4, 5, 7, 8, 10, 11, 17, 18, 24 and 25.

11. An isolated nucleic acid molecule a. comprising a nucleotide sequence encoding the polypeptide of any one of the preceding embodiments; or b. comprising a nucleotide sequence selected from SEQ ID NOs: 1, 2, 4, 5, 7, 8, 10, 11, 17, 18, 24, and 25, or a nucleotide sequence having at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to the nucleotide sequence of SEQ ID NOs: 1, 2, 4, 5, 7, 8, 17, 18, or 24, 25 and encoding a polypeptide of the HAD-like hydrolase superfamily, comprising terpene synthase activity, and in particular comprising the ability to produce a drimane sesquiterpene alcohol, and/or a phosphate precursor thereof, like a monophosphate and more particularly a diphosphate precursor thereof, in particular a drimenyl phosphate precursor like a monophosphate and more particularly an drimenyl diphosphate; and/oran albicanyl phosphate precursor like a monophosphate and more particularly an albicanyl diphosphate from famesyl diphosphate (FPP) as substrate; more particularly a product profile as described in Table 1, above; or c. comprising a nucleotide sequence comprising a sequence complementary to one of the sequences of b.; or d. comprising a nucleotide sequence hybridizing under stringent conditions to a nucleotide sequence of a., b. or c.

In one particular embodiment, the nucleic acid can be either present naturally in plants of the genus Bazzania or Selaginella, like the species Bazzania trilobata or Selaginella moellendorjfii, or in other plant species, or be obtained by modifying SEQ ID NOs: 1, 2, 4, 5, 7, 8, 10, 11, 17, 18, 24, and 25 or the reverse complement thereof.

In another embodiment, the nucleic acid is isolated or is derived from plants of the genus Bazzania or Selaginella, like the species Bazzania trilobata or Selaginella moellendorjfii. 12. An expression construct comprising at least one nucleic acid molecule of embodiment 11, optionally in combination with at least one regulatory sequence.

13. A vector comprising at least one nucleic acid molecule of embodiment 11 or at least one expression construct of embodiment 12.

14. The vector of embodiment 13, wherein the vector is a prokaryotic, viral or eukaryotic vector.

15. The vector of embodiment 13 or 14, wherein the vector is an expression vector.

16. The vector of anyone of the embodiments 13 to 15, which is a plasmid vector.

17. A recombinant, non-natural non-human host cell or a recombinant non-human host organism, prepared by genetic engineering, comprising a. at least one isolated nucleic acid molecule of embodiment 11, optionally stably integrated into the genome; or b. at least one expression construct of embodiment 12, optionally stably integrated into the genome; or c. at least one vector of any one of embodiments 13 to 16.

A recombinant non-human host cell or a recombinant non-human host organism of this embodiment differs from natural non-human host cells or non-human host organisms in that foreign genetic material such as an isolated nucleic acid, an expression construct or a vector is artificially introduced into the host organism or host cell. A recombinant non-human host cell or a recombinant non-human host organism of this embodiment is a non-human host cell or non-human host organism (genetically) modified to comprise the desired foreign genetic material such as an isolated nucleic acid, an expression construct or a vector as described in the embodiments above. In particular, the recombinant non-human host cell or the recombinant non-human host organism of this embodiment is a cell or organism different from the cell or organism in which the desired genetic material is naturally present. 18. The host cell or host organism of embodiment 17, selected from a prokaryotic or eukaryotic organism, or a cell derived therefrom.

19. The host cell or host organism of embodiment 18, selected from bacterial, fungal and plant cells or plants.

20. The host cell or host organism of embodiment 19, wherein said fungal cells are yeast cells.

21. The host cell or host organism of embodiment 20, wherein said yeast cells are selected from the genus Saccharomyces or Pichia, in particular from the species Saccharomyces cerevisiae or Pichia pastoris.

22. The host cell or host organism of embodiment 18, wherein said bacterial cells are selected from the genus Escherichia , in particular from the species E. coli.

Some of these host cells or host organisms do not produce FPP naturally. To be suitable to carry out the method of an embodiment as described herein, organisms or cells that do not produce an acyclic terpene pyrophosphate precursor, e.g. FPP, naturally are genetically modified, in particular by transformation, transduction or conjugation, more particularly by transformation, to produce said precursor. The non-human host organism or non-human host cell can be, for example, modified (e.g., transformed) either before the modification with the nucleic acid described according to any of the above embodiments or simultaneously. Methods to modify (e.g., transform) organisms so that they produce an acyclic terpene pyrophosphate precursor, e.g. FPP, are already known in the art. For example, introducing enzyme activities of the mevalonate pathway (isoprenoid pathway) or MEP pathway is a suitable strategy to make the organism produce FPP (see also the examples section herein).

23. A method for producing at least one catalytically active polypeptide according to any one of embodiments 1 to 10 comprising: a. culturing a non-human host cell or non-human host organism of embodiment 17 to express at least one polypeptide according to anyone of embodiments 1 to 10; and b. optionally isolating the polypeptide from the non-human host cell or organism cultured in step a. 24. The method of embodiment 23, further comprising, prior to step a. genetically modifying a non-human host cell or non-human host organism by inserting at least one nucleic acid according of embodiment 11, at least one expression construct of embodiment 12, or at least one vector of anyone of embodiments 13 to 16 into said non-human host cell or non-human host organism, in particular by transformation, transduction or conjugation, so that it expresses the polypeptide according to any one of embodiments 1 to 10.

25. A method for producing a drimane sesquiterpene alcohol, in particular drimenol and/or albicanol, comprising: a. contacting farnesyl diphosphate (FPP) with a polypeptide as defined in anyone of embodiments 1 to 10, or with a polypeptide prepared according to embodiment 23 or 24, thereby obtaining at least one phosphate, in particular a diphosphate of drimane sesquiterpene alcohol, in particular a drimenyl and/or albicanyl phosphate, more particularly drimenyl diphosphate and/or albicanyl diphosphate; b. chemically or enzymatically cleaving the phosphate moiety, in particular diphosphate moiety of said product obtained in step a.; and c. optionally isolating the drimane sesquiterpene alcohol, in particular drimenol and/or albicanol.

In a particular embodiment, the phosphate moiety is cleaved enzymatically, by applying a phosphatase enzyme, more particularly an acid or alkaline phosphatase. Alkaline phosphatases of different origin, like bacterial enzymes, are preferred. Suitable phosphatases are commercially available enzymes.

In one embodiment, the drimane sesquiterpene alcohol, in particular drimenol and/or albicanol, or the mixture comprising the drimane sesquiterpene alcohol is isolated.

In a particular embodiment, the above method further comprises as step d. processing of the drimane sesquiterpene alcohol, in particular drimenol and/or albicanol, formed in step b. or isolated in step c. to obtain a derivative thereof using chemical synthesis or biocatalytic synthesis (such as biochemical synthesis with expressed enzymes, or bioconversion with living cells that express enzymes) or a combination of both. Said drimane sesquiterpene alcohol derivative may in particular be selected from a hydrocarbon, alcohol, diol, triol, acetal, ketal, aldehyde, acid, ether, amide, ketone, lactone, epoxide, acetate, glycoside, and/or an ester. In one embodiment, the above method comprises contacting the drimane sesquiterpene alcohol with at least one enzyme to produce a drimane sesquiterpene alcohol derivative. In particular, the drimane sesquiterpene alcohol derivative can be obtained using a biochemical method by contacting the drimane sesquiterpene alcohol with an enzyme such as, but not limited to, an oxidoreductase, a monooxygenase, a dioxygenase, a transferase. The biochemical conversion can be performed in vitro using isolated enzymes, enzymes from lysed cells or in vivo using whole cells. In another embodiment, the above method comprises converting the drimane sesquiterpene alcohol to a drimane sesquiterpene alcohol derivative using chemical synthesis. In particular, the drimane sesquiterpene alcohol derivative can be obtained by a chemical method such as, but not limited to, oxidation, reduction, alkylation, acylation and/or rearrangement. In another embodiment, the above method comprises as step e. optionally isolating the derivative of step d.

26. The method of embodiment 25, wherein the drimane sesquiterpene alcohol comprises drimenol and/or albicanol, particularly as the main product.

27. The method of any one of embodiments 25 and 26, which comprises providing, in particular transforming, a non-human host cell or non-human host organism with at least one nucleic acid according to embodiment 11, at least one expression construct of embodiment 12, or at least one vector of anyone of the embodiments 13 to 16 so that it expresses a polypeptide according to any one of embodiments 1 to 10.

28. The method of any one of embodiments 25 to 27, wherein FPP is contacted with a non-human host cell or non-human host organism of any one of embodiments 17 to 22, with a cell lysate thereof, or a culture medium containing said non-human host cell or non-human host organism; and/or with the polypeptide as defined in anyone of the embodiments 1 to 10, expressed in or isolated from said non-human host cell or non-human host organism, cell lysate or culture medium.

29. The method of embodiment 28, wherein said drimane sesquiterpene alcohol is fermentatively produced by said non-human host organism or non-human host cell.

30. The method of embodiment 28, wherein said drimane sesquiterpene alcohol is produced by an enzymatic process, comprising the conversion of FPP with an isolated polypeptide of anyone of the embodiments 1 to 10, optionally in the presence of further adjuvants.

In a particular embodiment, in the method of any one of embodiments 25 to 30, the polypeptide comprises a. an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 3 or SEQ ID NO: 12; b. a class II synthase motif having the amino acid sequence PDDLDSTS (SEQ ID NO: 47); and c. a QW motif having the amino acid sequence QNVDGSW (SEQ ID NO: 52); and d. optionally at least one further sequence motif selected from the amino acid sequences i. Lxxxx(W/F)xxYxxG (SEQ ID NO: 56), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence LRSHIWFNYSMG (SEQ ID NO: 57); ii. YxDxxRxRVD(P/A)V(V/A)xxN (SEQ ID NO: 62), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence YFDPLRLRVDP V A ATN (SEQ ID NO: 63); and iii. GTx(Y/F)xYxxxExFL(Y/F) (SEQ ID NO: 69), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence GTLYYRTPEAFLY (SEQ ID NO: 70); and the drimane sesquiterpene alcohol comprises drimenol.

In another particular embodiment, in the method of any one of embodiments 25 to 30, the polypeptide comprises a. an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 6 or SEQ ID NO: 9; b. a class II synthase motif having the amino acid sequence PxDxD(T/S)(T/M)S (SEQ ID NO 46), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence PDDLDTTS (SEQ ID NO: 48) or PNDLDTTS (SEQ ID NO: 50); and c. a QW motif comprising the amino acid sequence QX 1X2 DGX 3 W (SEQ ID NO: 51), wherein xi and X 2 can be any naturally occurring amino acid residue and X 3 is G, and particularly the amino acid sequence QCDDGGW (SEQ ID NO: 53) or QSSDGGW (SEQ ID NO: 54); and d. optionally at least one further sequence motif selected from the amino acid sequences i. LRxxTWxxYECG (SEQ ID NO: 61), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence LRSATWAAYECG (SEQ ID NO: 58) or LRTPT W GKYEC G (SEQ ID NO: 59); ii. Y(F/L)Dx(T/E)RPRVD(P/A)VVx(A/V)N (SEQ ID NO: 68), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence YFDETRPRVDAVVNVN (SEQ ID NO: 64) or YFDKTRPRVDPVVCVN (SEQ ID NO: 65); and iii. GTx(Y/F)YxxxExFL(Y/F) (SEQ ID NO: 69), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence GTLFYYHAESFLY (SEQ ID NO: 71); and the drimane sesquiterpene alcohol comprises albicanol.

In another particular embodiment, in the method of any one of embodiments 25 to 30, the polypeptide comprises a. an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 19 or

SEQ ID NO: 26; b. a class II synthase motif having the amino acid sequence PPDIDTMS (SEQ ID NO:

49); and c. a QW motif having the amino acid sequence QNEDGSW (SEQ ID NO: 55); and d. optionally at least one further sequence motif selected from the amino acid sequences i. Lxxxx(W/F)xxYxxG (SEQ ID NO: 56), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence LQHSSFLAYSCG (SEQ ID NO: 60); ii. Y(F/L)Dx(T/E)RPRVD(P/A)VVx(A/V)N (SEQ ID NO: 68), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence YLDVERPRVDPVVIAN (SEQ ID NO: 66) or YLDLERPRVDPVVIAN (SEQ ID NO: 67); and iii. GTx(Y/F)xYxxxExFLx (SEQ ID NO: 69), wherein x can be any naturally occurring amino acid residue, and particularly the amino acid sequence GTRY YLS QEDFLF (SEQ ID NO: 72); and the drimane sesquiterpene alcohol comprises drimenol and albicanol. More particularly, the TPS as applied comprises a class II motif, a QW motif, and at least one of the conserved motifs L, ii. and iii..

Even more particularly, a TPS as applied comprises a class II motif, a QW motif, and at least two of the conserved motifs L, ii. and iii..

Still more particularly, a TPS as applied comprises a class II motif, a QW motif, and all three of the conserved motifs L, ii. and iii..

31. The use of a polypeptide as defined in anyone of the embodiments 1 to 10 for preparing odorants, flavours or fragrance ingredients, in particular Ambrox (preferably via drimenol/drimenyl diphosphate).

32. The use of a drimane sesquiterpene alcohol as prepared according to anyone of the embodiments 25 to 30 for preparing odorants, flavours or fragrance ingredients, in particular Ambrox.

33. A method of producing Ambrox, which method comprises a. providing drimenol and/or albicanol by a method of anyone of the embodiments as recited in any one of embodiments 25 to 30, b. optionally isolating drimenol and/or albicanol as produced in step a.; and c. converting drimenol and/or albicanol in a manner know per se to Ambrox, for example as reported in Tetrahedron: Asymmetry 11 (2000) 1375-1388.

34. A composition comprising the substance prepared according to embodiment 32 or 33.

35. The composition according to embodiment 34 selected from the group consisting of body care compositions, home care compositions and fragrance compositions.

36. A method for producing a drimane sesquiterpene alcohol, in particular drimenol and/or albicanol, comprising: a. culturing a non-human host organism or a non-human host cell capable of producing FPP and transformed to express a polypeptide of anyone of the embodiments 1 to 10; and b. optionally isolating the drimane sesquiterpene alcohol , in particular drimenol and/or albicanol. In one embodiment, a non-human host organism or a non-human host cell which does not produce FPP naturally is genetically modified, in particular by transformation, transduction or conjugation, more particularly by transformation, to produce said precursor. The non-human host organism or non-human host cell may be, for example, modified (e.g., transformed) either before the modification with the nucleic acid described according to any of the above embodiments or simultaneously. Methods to modify (e.g., transform) organisms so that they produce an acyclic terpene pyrophosphate precursor, e.g. FPP, are already known in the art. For example, introducing enzyme activities of the mevalonate pathway (isoprenoid pathway) or MEP pathway is a suitable strategy to make the organism produce FPP (see also the examples section herein).

37. A method for preparing a mutant polypeptide of the Haloacid dehalogenase-like (HAD-like) hydrolase superfamily comprising terpene synthase activity, in particular comprising the ability to produce a drimane sesquiterpene alcohol , and/or a phosphate derivative thereof, like a monophosphate and more particularly a diphosphate derivative thereof; and in particular to produce a drimenyl phosphate derivative like a monophosphate and more particularly a drimenyl diphosphate, and/or an albicanyl phosphate derivative like a monophosphate and more particularly an albicanyl diphosphate, from farnesyl diphosphate (FPP) as substrate, which method comprises the steps of: a. selecting a nucleic acid molecule according to embodiment 11; b. modifying the selected nucleic acid molecule to obtain at least one mutant nucleic acid molecule; c. genetically modifying non-human host cells or unicellular non-human host organisms with the mutant nucleic acid sequence to express a polypeptide encoded by the mutant nucleic acid sequence, in particular by horizontal gen transfer such as transformation, transduction or conjugation, more particularly by transforming said host cells or unicellular host organisms; d. screening the expression product for at least one mutant comprising terpene synthase activity, in particular comprising the ability to produce a drimane sesquiterpene alcohol , and/or a phosphate precursor thereof, like a monophosphate and more particularly a diphosphate derivative thereof; and in particular to produce a drimenyl phosphate precursor like a monophosphate and more particularly a drimenyl diphosphate, and/or an albicanyl phosphate precursor like a monophosphate and more particularly an albicanyl diphosphate, from FPP as substrate; and, e. optionally, if the polypeptide has no desired mutant activity, repeat the process steps a. to d. until a polypeptide with a desired mutant activity is obtained; and, f. optionally, if a polypeptide having a desired mutant activity was identified in step d., isolating the corresponding mutant nucleic acid obtained in step c.

If not stated otherwise within the above degree ranges of “at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity”, particular values are those of at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity and values of at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity are even more particular. c. Polypeptides applicable according to the invention

In this context the following definitions apply:

The generic term “polypeptide” or “peptide”, which may be used interchangeably, refers to a natural or synthetic linear chain of consecutive, peptidically linked amino acid residues (amino acid sequence), comprising about 10 residues up to more than 1.000 residues. In some embodiments provided herein, a polypeptide comprises an amino acid sequence that is an enzyme, or a fragment, or a variant thereof. Short chain polypeptides with up to 30 residues are also designated as “oligopeptides”.

The term “isolated polypeptide” refers to an amino acid sequence that is removed from its natural environment by any method or combination of methods known in the art and includes recombinant, biochemical and synthetic methods.

The term “protein” refers to a macromolecular structure consisting of one or more polypeptides. It includes oligopeptides, peptides, polypeptides and full length proteins whether naturally occurring or synthetic. The amino acid sequence of its polypeptide(s) represents the “primary structure” of the protein. The amino acid sequence also predetermines the “secondary structure” of the protein by the formation of special structural elements, such as alpha-helical and beta-sheet structures formed within a polypeptide chain. The arrangement of a plurality of such secondary structural elements defines the “tertiary structure” or spatial arrangement of the protein. If a protein comprises more than one polypeptide chains said chains are spatially arranged forming the “quaternary structure” of the protein. A correct spatial arrangement or “folding” of the protein is prerequisite of protein function. Denaturation or unfolding destroys protein function. If such destruction is reversible, protein function may be restored by refolding.

A typical protein function referred to herein is an “enzyme function”, i.e. the protein acts as biocatalyst on a substrate, for example a chemical compound, and catalyzes the conversion of said substrate to a product. An enzyme may show a high or low degree of substrate and/or product specificity.

A “polypeptide” referred to herein as having a particular “activity” thus implicitly refers to a correctly folded protein showing the indicated activity, as for example a specific enzyme activity. Thus, unless otherwise indicated the term “polypeptide” also encompasses the terms “protein” and “enzyme”.

“Target peptide” refers to an amino acid sequence which targets a protein, or polypeptide to intracellular organelles, i.e., mitochondria, or plastids, or to the extracellular space (secretion signal peptide). A nucleic acid sequence encoding a target peptide may be fused to the nucleic acid sequence encoding the amino terminal end, i.e., N-terminal end, of the protein or polypeptide, or may be used to replace a native targeting polypeptide.

The present invention also relates to "functional equivalents" (also designated as “analogs” or “functional mutations”) of the polypeptides specifically described herein.

For example, "functional equivalents" refer to polypeptides which, in a test used for determining enzymatic drimenyl and/or albicanyl diphosphate synthase activity, display at least a 1 to 10%, or at least 20%, or at least 50%, or at least 75%, or at least 90% higher or lower drimenyl and/or albicanyl diphosphate synthase activity, as that of the polypeptides specifically described herein.

"Functional equivalents”, according to the invention, also cover particular mutants, which, in at least one sequence position of an amino acid sequences stated herein, have an amino acid that is different from that concretely stated one, but nevertheless possess one of the aforementioned biological activities, as for example enzyme activity. "Functional equivalents" thus comprise mutants obtainable by one or more, like 1 to 20, in particular 1 to 15 or 5 to 10 amino acid additions, substitutions, in particular conservative substitutions (i.e. the amino acid in question is replaced by an amino acid of the same charge, size, polarity and/or solubility), deletions and/or inversions, where the stated changes can occur in any sequence position, provided they lead to a mutant with the profile of properties according to the invention. Functional equivalence is in particular also provided if the activity patterns coincide qualitatively between the mutant and the unchanged polypeptide, i.e. if, for example, interaction with the same agonist or antagonist or substrate, however at a different rate, (i.e. expressed by a EC50 or IC50 value or any other parameter suitable in the present technical field) is observed. Examples of suitable (conservative) amino acid substitutions are shown in the following table:

Original residue Examples of substitution

Ala Ser

Arg Lys

Asn Gin; His

Asp Glu

Cys Ser

Gin Asn

Glu Asp

Gly Pro

His Asn ; Gin lie Leu; Val

Leu lie; Val

Lys Arg ; Gin ; Glu

Met Leu ; He

Phe Met ; Leu ; Tyr

Ser Thr

Thr Ser

Trp Tyr

Tyr Trp ; Phe

Val He; Leu

“Functional equivalents” in the above sense are also “precursors” of the polypeptides described herein, as well as “functional derivatives” and “salts” of the polypeptides.

“Precursors” are in that case natural or synthetic precursors of the polypeptides with or without the desired biological activity. The expression “salts” means salts of carboxyl groups as well as salts of acid addition of amino groups of the protein molecules according to the invention. Salts of carboxyl groups can be produced in a known way and comprise inorganic salts, for example sodium, calcium, ammonium, iron and zinc salts, and salts with organic bases, for example amines, such as triethanolamine, arginine, lysine, piperidine and the like. Salts of acid addition, for example salts with inorganic acids, such as hydrochloric acid or sulfuric acid and salts with organic acids, such as acetic acid and oxalic acid, are also covered by the invention.

“Functional derivatives” of polypeptides according to the invention can also be produced on functional amino acid side groups or at their N-terminal or C-terminal end using known techniques. Such derivatives comprise for example aliphatic esters of carboxylic acid groups, amides of carboxylic acid groups, obtainable by reaction with ammonia or with a primary or secondary amine; N-acyl derivatives of free amino groups, produced by reaction with acyl groups; or O-acyl derivatives of free hydroxyl groups, produced by reaction with acyl groups.

’’Functional equivalents” naturally also comprise polypeptides that can be obtained from other organisms, as well as naturally occurring variants. For example, areas of homologous sequence regions can be established by sequence comparison, and equivalent polypeptides can be determined on the basis of the concrete parameters of the invention.

“Functional equivalents” also comprise “fragments”, like individual domains or sequence motifs, of the polypeptides according to the invention, or N- and or C-terminally truncated forms, which may or may not display the desired biological function. Preferably such “fragments” retain the desired biological function at least qualitatively.

“Functional equivalents” are, moreover, fusion proteins which have one of the polypeptide sequences stated herein or functional equivalents derived therefrom and at least one further, functionally different, heterologous sequence in functional N-terminal or C- terminal association (i.e. without substantial mutual functional impairment of the fusion protein parts). Non-limiting examples of these heterologous sequences are, e.g., signal peptides, histidine anchors or enzymes.

“Functional equivalents” which are also comprised in accordance with the invention are homologs to the specifically disclosed polypeptides. These have at least 60%, preferably at least 75%, in particular at least 80 or 85%, such as, for example, 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99%, homology (or identity) to one of the specifically disclosed amino acid sequences, calculated by the algorithm of Pearson and Lipman, Proc. Natl. Acad, Sci. (USA) 85(8), 1988, 2444-2448. A homology or identity, expressed as a percentage, of a homologous polypeptide according to the invention means in particular an identity, expressed as a percentage, of the amino acid residues based on the total length of one of the amino acid sequences described specifically herein.

The identity data, expressed as a percentage, may also be determined with the aid of BLAST alignments, algorithm blastp (protein-protein BLAST), or by applying the Clustal settings specified herein below.

In the case of a possible protein glycosylation, “functional equivalents” according to the invention comprise polypeptides as described herein in deglycosylated or glycosylated form as well as modified forms that can be obtained by altering the glycosylation pattern.

Functional equivalents or homologues of the polypeptides according to the invention can be produced by mutagenesis, e.g. by point mutation, lengthening or shortening of the protein or as described in more detail below. Functional equivalents or homologs of the polypeptides according to the invention can be identified by screening combinatorial databases of mutants, for example shortening mutants. For example, a variegated database of protein variants can be produced by combinatorial mutagenesis at the nucleic acid level, e.g. by enzymatic ligation of a mixture of synthetic oligonucleotides. There are a great many methods that can be used for the production of databases of potential homologues from a degenerated oligonucleotide sequence. Chemical synthesis of a degenerated gene sequence can be carried out in an automatic DNA synthesizer, and the synthetic gene can then be ligated in a suitable expression vector. The use of a degenerated genome makes it possible to supply all sequences in a mixture, which code for the desired set of potential protein sequences. Methods of synthesis of degenerated oligonucleotides are known to a person skilled in the art.

In the prior art, several techniques are known for the screening of gene products of combinatorial databases, which were produced by point mutations or shortening, and for the screening of cDNA libraries for gene products with a selected property. These techniques can be adapted for the rapid screening of the gene banks that were produced by combinatorial mutagenesis of homologues according to the invention. The techniques most frequently used for the screening of large gene banks, which are based on a high-throughput analysis, comprise cloning of the gene bank in expression vectors that can be replicated, transformation of the suitable cells with the resultant vector database and expression of the combinatorial genes in conditions in which detection of the desired activity facilitates isolation of the vector that codes for the gene whose product was detected. Recursive Ensemble Mutagenesis (REM), a technique that increases the frequency of functional mutants in the databases, can be used in combination with the screening tests, in order to identify homologues.

An embodiment provided herein provides orthologs and paralogs of polypeptides disclosed herein as well as methods for identifying and isolating such orthologs and paralogs. A definition of the terms “ortholog” and “paralog” is given below and applies to amino acid and nucleic acid sequences.

The polypeptides of the invention include all active forms, including active subsequences, e.g., catalytic domains or active sites, of an enzyme of the invention. In one aspect, the invention provides catalytic domains or active sites as set forth below. In one aspect, the invention provides a peptide or polypeptide comprising or consisting of an active site domain as predicted through use of a database such as Pfam (http://pfam.wustl.edu/hmmsearch.shtml) (which is a large collection of multiple sequence alignments and hidden Markov models covering many common protein families, The Pfam protein families database, A. Bateman, E. Birney, L. Cerruti, R. Durbin, L. Etwiller, S. R. Eddy, S. Griffiths-Jones, K. L. Howe, M. Marshall, and E. L. L. Sonnhammer, Nucleic Acids Research, 30(l):276-280, 2002) or equivalent, as for example InterPro and SMART databases (http://www.ebi.ac.uk/interpro/scan.html, http://smart.embl-heidelberg.de/).

The invention also encompasses a “polypeptide variant” having the desired activity, wherein the variant polypeptide is selected from an amino acid sequence having at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%, sequence identity to a specific, in particular natural, amino acid sequence as referred to by a specific SEQ ID NO and contains at least one, as for example 1 to 30 or 1 to 20, like 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid sequence differences, such as amino acid additions, substitutions, insertions or deletions relative to said (non-modified) SEQ ID NO. d. Coding nucleic acid sequences applicable according to the invention

In this context the following definitions apply:

The terms “nucleic acid sequence”, “nucleic acid”, “nucleic acid molecule” and “polynucleotide” are used interchangeably meaning a sequence of nucleotides. A nucleic acid sequence may be a single-stranded or double-stranded deoxyribonucleotide, or ribonucleotide, of any length, and include coding and non-coding sequences of a gene, exons, introns, sense and anti-sense complimentary sequences, genomic DNA, cDNA, miRNA, siRNA, mRNA, rRNA, tRNA, recombinant nucleic acid sequences, isolated and purified naturally occurring DNA and/or RNA sequences, synthetic DNA and RNA sequences, fragments, primers and nucleic acid probes. The skilled artisan is aware that the nucleic acid sequences of RNA are identical to the DNA sequences with the difference of thymine (T) being replaced by uracil (U). The term “nucleotide sequence” should also be understood as comprising a polynucleotide molecule or an oligonucleotide molecule in the form of a separate fragment or as a component of a larger nucleic acid.

The term “naturally-occurring” as used herein as applied to a nucleic acid refers to a nucleic acid that is found in a cell or an organism in nature and which has not been intentionally modified by a human in the laboratory.

A “fragment” of a polynucleotide or nucleic acid sequence refers to a contiguous nucleotide that is particularly at least 15 bp, at least 30 bp, at least 40 bp, at least 50 bp and/or at least 60 bp in length of the polynucleotide of an embodiment herein. Particularly, the fragment of a polynucleotide comprises at least 25, more particularly at least 50, more particularly at least 75, more particularly at least 100, more particularly at least 150, more particularly at least 200, more particularly at least 300, more particularly at least 400, more particularly at least 500, more particularly at least 600, more particularly at least 700, more particularly at least 800, more particularly at least 900, more particularly at least 1000 contiguous nucleotides of the polynucleotide of an embodiment herein. Without being limited, the fragment of the polynucleotides herein may be used as a PCR primer, and/or as a probe, or for anti-sense gene silencing or RNAi.

“Recombinant nucleic acid sequences” are nucleic acid sequences that result from the use of laboratory methods (for example, molecular cloning) to bring together genetic material from more than one source, creating or modifying a nucleic acid sequence that does not occur naturally and would not be otherwise found in biological organisms.

“Recombinant DNA technology” refers to molecular biology procedures to prepare a recombinant nucleic acid sequence as described, for instance, in Laboratory Manuals edited by Weigel and Glazebrook, 2002, Cold Spring Harbor Lab Press; and Sambrook et ah, 1989, Cold Spring Harbor, NY, Cold Spring Harbor Laboratory Press.

The term “gene” means a DNA sequence comprising a region which is transcribed into a RNA molecule, e.g., an mRNA in a cell, operably linked to suitable regulatory regions, e.g., a promoter. A gene may thus comprise several operably linked sequences, such as a promoter, a 5'-leader sequence comprising, e.g., sequences involved in translation initiation, a coding region of cDNA or genomic DNA, introns, exons, and/or a 3'-non-translated sequence comprising, e.g., transcription termination sites.

A “chimeric gene” refers to any gene which is not normally found in nature in a species, in particular, a gene in which one or more parts of the nucleic acid sequence are present that are not associated with each other in nature. For example the promoter is not associated in nature with part or all of the transcribed region or with another regulatory region. The term “chimeric gene” is understood to include expression constructs in which a promoter or transcription regulatory sequence is operably linked to one or more coding sequences or to an antisense, i.e., reverse complement of the sense strand, or inverted repeat sequence (sense and antisense, whereby the RNA transcript forms double stranded RNA upon transcription). The term “chimeric gene” also includes genes obtained through the combination of portions of one or more coding sequences to produce a new gene.

A “3' UTR” or “3' non-translated sequence” (also referred to as “3' untranslated region”, or “3'end”) refers to the nucleic acid sequence found downstream of the coding sequence of a gene, which comprises, for example, a transcription termination site and (in most, but not all eukaryotic mRNAs) a polyadenylation signal such as AAUAAA or variants thereof. After termination of transcription, the mRNA transcript may be cleaved downstream of the polyadenylation signal and a poly(A) tail may be added, which is involved in the transport of the mRNA to the site of translation, e.g., the cytoplasm.

The term “primer” refers to a short nucleic acid sequence that is hybridized to a template nucleic acid sequence and is used for polymerization of a nucleic acid sequence complementary to the template.

The invention also relates to nucleic acid sequences that code for polypeptides as defined herein.

In particular, the invention also relates to nucleic acid sequences (single-stranded and double- stranded DNA and RNA sequences, e.g. cDNA, genomic DNA and mRNA), coding for one of the above polypeptides and their functional equivalents which can be obtained for example using artificial nucleotide analogs.

The present invention also relates to nucleic acids with a certain degree of “identity” to the sequences specifically disclosed herein. “Identity” between two nucleic acids means identity of the nucleotides, in each case over the entire length of the nucleic acid.

The “identity” between two nucleotide sequences (the same applies to peptide or amino acid sequences) is a function of the number of nucleotide residues (or amino acid residues) that are identical in the two sequences when an alignment of these two sequences has been generated. Identical residues are defined as residues that are the same in the two sequences in a given position of the alignment. The percentage of sequence identity, as used herein, is calculated from the optimal alignment by taking the number of residues identical between two sequences dividing it by the total number of residues in the shortest sequence and multiplying by 100. The optimal alignment is the alignment in which the percentage of identity is the highest possible. Gaps may be introduced into one or both sequences in one or more positions of the alignment to obtain the optimal alignment. These gaps are then taken into account as non-identical residues for the calculation of the percentage of sequence identity. Alignment for the purpose of determining the percentage of amino acid or nucleic acid sequence identity can be achieved in various ways using computer programs and for instance publicly available computer programs available on the world wide web. Particularly, the BLAST program (Tatiana et al, FEMS Microbiol Lett., 1999, 174:247-250, 1999) set to the default parameters, available from the National Center for Biotechnology Information (NCBI) website at https://blast.ncbi.nlm.nih.gov/Blast.cgi, can be used to obtain an optimal alignment of protein or nucleic acid sequences and to calculate the percentage of sequence identity. In another example the identity may be calculated by means of the Vector NTI Suite 7.1 program of the company Informax (USA) employing the Clustal Method (Higgins DG, Sharp PM. (1989)) with the following settings:

Multiple alignment parameters:

Gap opening penalty 10

Gap extension penalty 10

Gap separation penalty range 8

Gap separation penalty off

% identity for alignment delay 40

Residue specific gaps off

Hydrophilic residue gap off

Transition weighing 0

Pairwise alignment parameter:

FAST algorithm on

K-tuple size 1

Gap penalty 3

Window size 5

Number of best diagonals 5

Alternatively the identity may be determined according to Chenna, et al. (2003), the web page: http://www.ebi.ac.Uk/Tools/clustalw/index.html# and the following settings

DNA Gap Open Penalty 15.0

DNA Gap Extension Penalty 6.66

DNA Matrix Identity

Protein Gap Open Penalty 10.0

Protein Gap Extension Penalty 0.2

Protein matrix Gonnet

Protein/DNA ENDGAP -1

Protein/DNA GAPDIST 4

All the nucleic acid sequences mentioned herein (single-stranded and double-stranded DNA and RNA sequences, for example cDNA and mRNA) can be produced in a known way by chemical synthesis from the nucleotide building blocks, e.g. by fragment condensation of individual overlapping, complementary nucleic acid building blocks of the double helix. Chemical synthesis of oligonucleotides can, for example, be performed in a known way, by the phosphoamidite method (Voet, Voet, 2nd edition, Wiley Press, New York, pages 896- 897). The accumulation of synthetic oligonucleotides and filling of gaps by means of the Klenow fragment of DNA polymerase and ligation reactions as well as general cloning techniques are described in Sambrook et al. (1989), see below. The nucleic acid molecules according to the invention can in addition contain non- translated sequences from the 3' and/or 5' end of the coding genetic region.

The invention further relates to the nucleic acid molecules that are complementary to the concretely described nucleotide sequences or a segment thereof.

The nucleotide sequences according to the invention make possible the production of probes and primers that can be used for the identification and/or cloning of homologous sequences in other cellular types and organisms. Such probes or primers generally comprise a nucleotide sequence region which hybridizes under "stringent" conditions (as defined herein elsewhere) on at least about 12, preferably at least about 25, for example about 40, 50 or 75 successive nucleotides of a sense strand of a nucleic acid sequence according to the invention or of a corresponding antisense strand.

“Homologous” sequences include orthologous or paralogous sequences. Methods of identifying orthologs or paralogs including phylogenetic methods, sequence similarity and hybridization methods are known in the art and are described herein.

“Paralogs” result from gene duplication that gives rise to two or more genes with similar sequences and similar functions. Paralogs typically cluster together and are formed by duplications of genes within related plant species. Paralogs are found in groups of similar genes using pair-wise Blast analysis or during phylogenetic analysis of gene families using programs such as CLUSTAL. In paralogs, consensus sequences can be identified characteristic to sequences within related genes and having similar functions of the genes.

“Orthologs”, or “orthologous sequences”, are sequences similar to each other because they are found in species that descended from a common ancestor. For instance, plant species that have common ancestors are known to contain many enzymes that have similar sequences and functions. The skilled artisan can identify orthologous sequences and predict the functions of the orthologs, for example, by constructing a polygenic tree for a gene family of one species using CLUSTAL or BLAST programs. A method for identifying or confirming similar functions among homologous sequences is comparing of the transcript profiles in host cells or host organisms, such as plants or microorganisms, overexpressing or lacking (in knockouts/knockdowns) related polypeptides. The skilled person will understand that genes having similar transcript profiles, with greater than 50% regulated transcripts in common, or with greater than 70% regulated transcripts in common, or greater than 90% regulated transcripts in common will have similar functions. Homologs, paralogs, orthologs and any other variants of the sequences herein are expected to function in a similar manner by making the host cells, host organisms such as plants or microorganisms producing terpene synthase proteins.

The term “selectable marker” refers to any gene which upon expression may be used to select a cell or cells that include the selectable marker. Examples of selectable markers are described below. The skilled artisan will know that different antibiotic, fungicide, auxotrophic or herbicide selectable markers are applicable to different target species.

The invention relates to both isolated nucleic acid molecules, which code for polypeptides according to the invention or biologically active segments thereof, and to nucleic acid fragments, which can be used for example as hybridization probes or primers for identifying or amplifying coding nucleic acids according to the invention.

An “isolated nucleic acid” or “isolated nucleic acid sequence” relates to a nucleic acid or nucleic acid sequence that is in an environment different from that in which the nucleic acid or nucleic acid sequence naturally occurs and can include those that are substantially free from contaminating endogenous material. An isolated nucleic acid molecule is, thus, separated from other nucleic acid molecules that are present in the natural source of the nucleic acid and can moreover be substantially free from other cellular material or culture medium, if it is being produced by recombinant techniques, or can be free from chemical precursors or other chemicals, if it is being synthesized chemically.

A nucleic acid molecule according to the invention can be isolated by means of standard techniques of molecular biology and the sequence information supplied according to the invention. For example, cDNA can be isolated from a suitable cDNA library, using one of the concretely disclosed complete sequences or a segment thereof as hybridization probe and standard hybridization techniques (as described for example in Sambrook, (1989)).

In addition, a nucleic acid molecule comprising one of the disclosed sequences, or a segment thereof, can be isolated by the polymerase chain reaction, using the oligonucleotide primers that were constructed on the basis of this sequence. The nucleic acid amplified in this way can be cloned in a suitable vector and can be characterized by DNA sequencing. The oligonucleotides according to the invention can also be produced by standard methods of synthesis, e.g. using an automatic DNA synthesizer.

Nucleic acid sequences according to the invention or derivatives thereof, homologues or parts of these sequences, can for example be isolated by usual hybridization techniques or the PCR technique from other bacteria, e.g. via genomic or cDNA libraries. These DNA sequences hybridize in standard conditions with the sequences according to the invention. “Hybridize" means the ability of a polynucleotide or oligonucleotide to bind to an almost complementary sequence in standard conditions, whereas nonspecific binding does not occur between non-complementary partners in these conditions. For this, the sequences can be 90-100% complementary. The property of complementary sequences of being able to bind specifically to one another is utilized for example in Northern Blotting or Southern Blotting or in primer binding in PCR or RT-PCR.

Short oligonucleotides of the conserved regions are used advantageously for hybridization. However, it is also possible to use longer fragments of the nucleic acids according to the invention or the complete sequences for the hybridization. These “standard conditions” vary depending on the nucleic acid used (oligonucleotide, longer fragment or complete sequence) or depending on which type of nucleic acid - DNA or RNA - is used for hybridization. For example, the melting temperatures for DNA:DNA hybrids are approx. 10°C lower than those of DNA:RNA hybrids of the same length.

For example, depending on the particular nucleic acid, standard conditions mean temperatures between 42 and 58°C in an aqueous buffer solution with a concentration between 0.1 to 5 x SSC (1 x SSC = 0.15 M NaCl, 15 mM sodium citrate, pH 7.2) or additionally in the presence of 50% formamide, for example 42°C in 5 x SSC, 50% formamide. Advantageously, the hybridization conditions for DNA:DNA hybrids are 0.1 x SSC and temperatures between about 20°C to 45°C, preferably between about 30°C to 45°C. For DNA:RNA hybrids the hybridization conditions are advantageously 0.1 x SSC and temperatures between about 30°C to 55°C, preferably between about 45°C to 55°C. These stated temperatures for hybridization are examples of calculated melting temperature values for a nucleic acid with a length of approx. 100 nucleotides and a G + C content of 50% in the absence of formamide. The experimental conditions for DNA hybridization are described in relevant genetics textbooks, for example Sambrook et ah, 1989, and can be calculated using formulae that are known by a person skilled in the art, for example depending on the length of the nucleic acids, the type of hybrids or the G + C content. A person skilled in the art can obtain further information on hybridization from the following textbooks: Ausubel et al. (eds), (1985), Brown (ed) (1991).

"Hybridization" can in particular be carried out under stringent conditions. Such hybridization conditions are for example described in Sambrook (1989), or in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6.

As used herein, the term “hybridization” or “hybridizes under certain conditions” is intended to describe conditions for hybridization and washes under which nucleotide sequences that are significantly identical or homologous to each other remain bound to each other. The conditions may be such that sequences, which are at least about 70%, such as at least about 80%, and such as at least about 85%, 90%, or 95% identical, remain bound to each other. Definitions of low stringency, moderate, and high stringency hybridization conditions are provided herein.

Appropriate hybridization conditions can be selected by those skilled in the art with minimal experimentation as exemplified in Ausubel el al. (1995, Current Protocols in Molecular Biology, John Wiley & Sons, sections 2, 4, and 6). Additionally, stringency conditions are described in Sambrook et al. (1989, Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Press, chapters 7, 9, and 11).

As used herein, defined conditions of low stringency are as follows. Filters containing DNA are pretreated for 6 h at 40°C in a solution containing 35% formamide, 5x SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0.1% PVP, 0.1% Ficoll, 1% BSA, and 500 pg/ml denatured salmon sperm DNA. Hybridizations are carried out in the same solution with the following modifications: 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 pg/ml salmon sperm DNA, 10% (wt/vol) dextran sulfate, and 5-20xl0 6 cpm of 32P-labeled probe is used. Filters are incubated in hybridization mixture for 18-20 h at 40°C, and then washed for 1.5 h at 55°C in a solution containing 2x SSC, 25 mM Tris-HCl (pH 7.4), 5 mM EDTA, and 0.1% SDS. The wash solution is replaced with fresh solution and incubated an additional 1.5 h at 60°C. Filters are blotted dry and exposed for autoradiography.

As used herein, defined conditions of moderate stringency are as follows. Filters containing DNA are pretreated for 7 h at 50°C in a solution containing 35% formamide, 5x SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0.1% PVP, 0.1% Ficoll, 1% BSA, and 500 pg/ml denatured salmon sperm DNA. Hybridizations are carried out in the same solution with the following modifications: 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 pg/ml salmon sperm DNA, 10% (wt/vol) dextran sulfate, and 5-20xl0 6 cpm of 32P-labeled probe is used. Filters are incubated in hybridization mixture for 30 h at 50°C, and then washed for 1.5 h at 55°C in a solution containing 2x SSC, 25 mM Tris-HCl (pH 7.4), 5 mM EDTA, and 0.1% SDS. The wash solution is replaced with fresh solution and incubated an additional 1.5 h at 60°C. Filters are blotted dry and exposed for autoradiography.

As used herein, defined conditions of high stringency are as follows. Prehybridization of filters containing DNA is carried out for 8 h overnight at 65°C in buffer composed of 6 x SSC, 50 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02% BSA, and 500 pg/ml denatured salmon sperm DNA. Filters are hybridized for 48 h at 65°C in the prehybridization mixture containing 100 pg /ml denatured salmon sperm DNA and 5-20xl0 6 cpm of 32P-labeled probe. Washing of filters is done at 37°C for 1 h in a solution containing 2x SSC, 0.01% PVP, 0.01% Ficoll, and 0.01% BSA. This is followed by a wash in O.lx SSC at 50°C for 45 minutes.

Other conditions of low, moderate, and high stringency well known in the art (e.g., as employed for cross-species hybridizations) may be used if the above conditions are inappropriate (e.g., as employed for cross-species hybridizations).

A detection kit for nucleic acid sequences encoding a polypeptide of the invention may include primers and/or probes specific for nucleic acid sequences encoding the polypeptide, and an associated protocol to use the primers and/or probes to detect nucleic acid sequences encoding the polypeptide in a sample. Such detection kits may be used to determine whether a plant, organism, microorganism or cell has been modified, i.e., transformed with a sequence encoding the polypeptide.

To test a function of variant DNA sequences according to an embodiment herein, the sequence of interest is operably linked to a selectable or screenable marker gene and expression of said reporter gene is tested in transient expression assays, for example, with microorganisms or with protoplasts or in stably transformed plants.

The invention also relates to derivatives of the concretely disclosed or derivable nucleic acid sequences.

Thus, further nucleic acid sequences according to the invention can be derived from the sequences specifically disclosed herein and can differ from it by one or more, like 1 to 20, in particular 1 to 15 or 5 to 10 additions, substitutions, insertions or deletions of one or several (like for example 1 to 10) nucleotides, and furthermore code for polypeptides with the desired profile of properties.

The invention also encompasses nucleic acid sequences that comprise so-called silent mutations or have been altered, in comparison with a concretely stated sequence, according to the codon usage of a special original or host organism.

According to a particular embodiment of the invention variant nucleic acids may be prepared in order to adapt its nucleotide sequence to a specific expression system. For example, bacterial expression systems are known to more efficiently express polypeptides if amino acids are encoded by particular codons. Due to the degeneracy of the genetic code, more than one codon may encode the same amino acid sequence, multiple nucleic acid sequences can code for the same protein or polypeptide, all these DNA sequences being encompassed by an embodiment herein. Where appropriate, the nucleic acid sequences encoding the polypeptides described herein may be optimized for increased expression in the host cell. For example, nucleic acids of an embodiment herein may be synthesized using codons particular to a host for improved expression.

The invention also encompasses naturally occurring variants, e.g. splicing variants or allelic variants, of the sequences described therein.

Allelic variants may have at least 60% homology at the level of the derived amino acid, preferably at least 80% homology, quite especially preferably at least 90% homology over the entire sequence range (regarding homology at the amino acid level, reference should be made to the details given above for the polypeptides). Advantageously, the homologies can be higher over partial regions of the sequences.

The invention also relates to sequences that can be obtained by conservative nucleotide substitutions (i.e. as a result thereof the amino acid in question is replaced by an amino acid of the same charge, size, polarity and/or solubility).

The invention also relates to the molecules derived from the concretely disclosed nucleic acids by sequence polymorphisms. Such genetic polymorphisms may exist in cells from different populations or within a population due to natural allelic variation. Allelic variants may also include functional equivalents. These natural variations usually produce a variance of 1 to 5% in the nucleotide sequence of a gene. Said polymorphisms may lead to changes in the amino acid sequence of the polypeptides disclosed herein. Allelic variants may also include functional equivalents.

Furthermore, derivatives are also to be understood to be homologs of the nucleic acid sequences according to the invention, for example animal, plant, fungal or bacterial homologs, shortened sequences, single-stranded DNA or RNA of the coding and noncoding DNA sequence. For example, homologs have, at the DNA level, a homology of at least 40%, preferably of at least 60%, especially preferably of at least 70%, quite especially preferably of at least 80% over the entire DNA region given in a sequence specifically disclosed herein.

Moreover, derivatives are to be understood to be, for example, fusions with promoters. The promoters that are added to the stated nucleotide sequences can be modified by at least one nucleotide exchange, at least one insertion, inversion and/or deletion, though without impairing the functionality or efficacy of the promoters. Moreover, the efficacy of the promoters can be increased by altering their sequence or can be exchanged completely with more effective promoters even of organisms of a different genus. e. Generation of functional polypeptide mutants Moreover, a person skilled in the art is familiar with methods for generating functional mutants, that is to say nucleotide sequences which code for a polypeptide with at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to anyone of amino acid related SEQ ID NOs as disclosed herein and/or encoded by a nucleic acid molecule comprising a nucleotide sequence having at least 70% sequence identity to anyone of the nucleotide related SEQ ID NOs as disclosed herein.

Depending on the technique used, a person skilled in the art can introduce entirely random or else more directed mutations into genes or else noncoding nucleic acid regions (which are for example important for regulating expression) and subsequently generate genetic libraries. The methods of molecular biology required for this purpose are known to the skilled worker and for example described in Sambrook and Russell, Molecular Cloning. 3 rd Edition, Cold Spring Harbor Laboratory Press 2001.

Methods for modifying genes and thus for modifying the polypeptide encoded by them have been known to the skilled worker for a long time, such as, for example

- site-specific mutagenesis, where individual or several nucleotides of a gene are replaced in a directed fashion (Trower MK (Ed.) 1996; In vitro mutagenesis protocols. Humana Press, New Jersey),

- saturation mutagenesis, in which a codon for any amino acid can be exchanged or added at any point of a gene (Kegler-Ebo DM, Docktor CM, DiMaio D (1994) Nucleic Acids Res 22:1593; Barettino D, Feigenbutz M, Valcarel R, Stunnenberg HG (1994) Nucleic Acids Res 22:541; Barik S (1995) Mol Biotechnol 3:1),

- error-prone polymerase chain reaction, where nucleotide sequences are mutated by error-prone DNA polymerases (Eckert KA, Kunkel TA (1990) Nucleic Acids Res 18:3739),

- the SeSaM method (sequence saturation method), in which preferred exchanges are prevented by the polymerase. Schenk et al., Biospektmm, Vol. 3, 2006, 277-279),

- the passaging of genes in mutator strains, in which, for example owing to defective DNA repair mechanisms, there is an increased mutation rate of nucleotide sequences (Greener A, Callahan M, Jerpseth B (1996) An efficient random mutagenesis technique using an E.coli mutator strain. In: Trower MK (Ed.) In vitro mutagenesis protocols. Humana Press, New Jersey), or

- DNA shuffling, in which a pool of closely related genes is formed and digested and the fragments are used as templates for a polymerase chain reaction in which, by repeated strand separation and reassociation, full-length mosaic genes are ultimately generated (Stemmer WPC (1994) Nature 370:389; Stemmer WPC (1994) Proc Natl Acad Sci USA 91:10747).

Using so-called directed evolution (described, inter alia, in Reetz MT and Jaeger K-E (1999), Topics Curr Chem 200:31; Zhao H, Moore JC, Volkov AA, Arnold FH (1999), Methods for optimizing industrial polypeptides by directed evolution, In: Demain AL, Davies JE (Ed.) Manual of industrial microbiology and biotechnology, American Society for Microbiology), a skilled worker can produce functional mutants in a directed manner and on a large scale. To this end, in a first step, gene libraries of the respective polypeptides are first produced, for example using the methods given above. The gene libraries are expressed in a suitable way, for example by bacteria or by phage display systems.

The relevant genes of host organisms which express functional mutants with properties that largely correspond to the desired properties can be submitted to another mutation cycle. The steps of the mutation and selection or screening can be repeated iteratively until the present functional mutants have the desired properties to a sufficient extent. Using this iterative procedure, a limited number of mutations, for example 1, 2, 3, 4 or 5 mutations, can be performed in stages and assessed and selected for their influence on the activity in question. The selected mutant can then be submitted to a further mutation step in the same way. In this way, the number of individual mutants to be investigated can be reduced significantly.

The results according to the invention also provide important information relating to structure and sequence of the relevant polypeptides, which is required for generating, in a targeted fashion, further polypeptides with desired modified properties. In particular, it is possible to define so-called “hot spots”, i.e. sequence segments that are potentially suitable for modifying a property by introducing targeted mutations.

Information can also be deduced regarding amino acid sequence positions, in the region of which mutations can be effected that should probably have little effect on the activity, and can be designated as potential “silent mutations”. f. Constructs for expressing polypeptides of the invention

In this context the following definitions apply:

“Expression of a gene” encompasses “heterologous expression” and “overexpression” and involves transcription of the gene and translation of the mRNA into a protein. Overexpression refers to the production of the gene product as measured by levels of mRNA, polypeptide and/or enzyme activity in transgenic cells or organisms that exceeds levels of production in non-transformed cells or organisms of a similar genetic background.

“Expression vector” as used herein means a nucleic acid molecule engineered using molecular biology methods and recombinant DNA technology for delivery of foreign or exogenous DNA into a host cell. The expression vector typically includes sequences required for proper transcription of the nucleotide sequence. The coding region usually codes for a protein of interest but may also code for an RNA, e.g., an antisense RNA, siRNA and the like.

An “expression vector” as used herein includes any linear or circular recombinant vector including but not limited to viral vectors, bacteriophages and plasmids. The skilled person is capable of selecting a suitable vector according to the expression system. In one embodiment, the expression vector includes the nucleic acid of an embodiment herein operably linked to at least one “regulatory sequence”, which controls transcription, translation, initiation and termination, such as a transcriptional promoter, operator or enhancer, or an mRNA ribosomal binding site and, optionally, including at least one selection marker. Nucleotide sequences are “operably linked” when the regulatory sequence functionally relates to the nucleic acid of an embodiment herein.

“Regulatory sequence” refers to a nucleic acid sequence that determines expression level of the nucleic acid sequences of an embodiment herein and is capable of regulating the rate of transcription of the nucleic acid sequence operably linked to the regulatory sequence. Regulatory sequences comprise promoters, enhancers, transcription factors, promoter elements and the like.

“Promoter” refers to a nucleic acid sequence that controls the expression of a coding sequence by providing a binding site for RNA polymerase and other factors required for proper transcription including, without limitation, transcription factor binding sites, repressor and activator protein binding sites. The meaning of the term promoter also includes the term “promoter regulatory sequence”. Promoter regulatory sequences may include upstream and downstream elements that may influences transcription, RNA processing or stability of the associated coding nucleic acid sequence. Promoters include naturally-derived and synthetic sequences. The coding nucleic acid sequence is usually located downstream of the promoter with respect to the direction of the transcription starting at the transcription initiation site.

In this context, a “functional” or “operative” linkage is understood as meaning for example the sequential arrangement of one of the nucleic acids with a regulatory sequence. For example the sequence with promoter activity and of a nucleic acid sequence to be transcribed and optionally further regulatory elements, for example nucleic acid sequences which ensure the transcription of nucleic acids, and for example a terminator, are linked in such a way that each of the regulatory elements can perform its function upon transcription of the nucleic acid sequence. This does not necessarily require a direct linkage in the chemical sense. Genetic control sequences, for example enhancer sequences, can even exert their function on the target sequence from more remote positions or even from other DNA molecules. Preferred arrangements are those in which the nucleic acid sequence to be transcribed is positioned behind (i.e. at the 3 ’-end of) the promoter sequence so that the two sequences are joined together covalently. The distance between the promoter sequence and the nucleic acid sequence to be expressed recombinantly can be smaller than 200 base pairs, or smaller than 100 base pairs or smaller than 50 base pairs.

In addition to promoters and terminator, the following may be mentioned as examples of other regulatory elements: targeting sequences, enhancers, polyadenylation signals, selectable markers, amplification signals, replication origins and the like. Suitable regulatory sequences are described, for example, in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990).

The term “constitutive promoter” refers to an unregulated promoter that allows for continual transcription of the nucleic acid sequence it is operably linked to.

As used herein, the term “operably linked” refers to a linkage of polynucleotide elements in a functional relationship. A nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For instance, a promoter, or rather a transcription regulatory sequence, is operably linked to a coding sequence if it affects the transcription of the coding sequence. Operably linked means that the DNA sequences to be linked are typically contiguous. The nucleotide sequence associated with the promoter sequence may be of homologous or heterologous origin with respect to the plant to be transformed. The sequence also may be entirely or partially synthetic. Regardless of the origin, the nucleic acid sequence associated with the promoter sequence will be expressed or silenced in accordance with promoter properties to which it is linked after binding to the polypeptide of an embodiment herein. The associated nucleic acid may code for a protein that is desired to be expressed or suppressed throughout the organism at all times or, alternatively, at a specific time or in specific tissues, cells, or cell compartments. Such nucleotide sequences particularly encode proteins conferring desirable phenotypic traits to the host cells or host organism altered (genetically modified) or transformed therewith. More particularly, the associated nucleotide sequence leads to the production of the product or products of interest as herein defined in the cell or organism, such as particularly albicanol and/or drimenol or a mixture comprising albicanol and/or drimenol or a mixture comprising albicanol and/or drimenol and one or more terpenes. Particularly, the nucleotide sequence encodes a polypeptide having an enzyme activity as herein defined, such as particularly a terpene synthase.

An “expression system” as used herein encompasses any combination of nucleic acid molecules required for the expression of one, or the co-expression of two or more polypeptides either in vivo of a given expression host, or in vitro. The respective coding sequences may either be located on a single nucleic acid molecule or vector, as for example a vector containing multiple cloning sites, or on a polycistronic nucleic acid, or may be distributed over two or more physically distinct vectors.

As used herein, the terms “amplifying” and “amplification” refer to the use of any suitable amplification methodology for generating or detecting recombinant of naturally expressed nucleic acid, as described in detail, below. For example, the invention provides methods and reagents (e.g., specific degenerate oligonucleotide primer pairs, oligo dT primer) for amplifying (e.g., by polymerase chain reaction, PCR) naturally expressed (e.g., genomic DNA or mRNA) or recombinant (e.g., cDNA) nucleic acids of the invention in vivo , ex vivo or in vitro.

The nucleotide sequence as described herein above may be part of an “expression cassette”. The terms “expression cassette” and “expression construct” are used synonymously. The (preferably recombinant) expression construct contains a nucleotide sequence which encodes a polypeptide according to the invention and which is under genetic control of regulatory nucleic acid sequences.

In a process applied according to the invention, the expression cassette may be part of an “expression vector”, in particular of a recombinant expression vector.

An “expression unit” is understood as meaning, in accordance with the invention, a nucleic acid with expression activity which comprises a promoter as defined herein and, after functional linkage with a nucleic acid to be expressed or with a gene to be expressed, regulates the expression, i.e. the transcription and the translation of said nucleic acid or said gene. It is therefore in this connection also referred to as a “regulatory nucleic acid sequence”. In addition to the promoter, other regulatory elements, for example enhancers, can also be present.

An “expression cassette” or “expression construct” is understood as meaning, in accordance with the invention, an expression unit which is functionally linked to the nucleic acid to be expressed or the gene to be expressed. In contrast to an expression unit, an expression cassette therefore comprises not only nucleic acid sequences which regulate transcription and translation, but also the nucleic acid sequences that are to be expressed as protein as a result of transcription and translation.

The terms “expression” or “overexpression” describe, in the context of the invention, the production or increase in intracellular activity of one or more polypeptides in a microorganism, which are encoded by the corresponding DNA. To this end, it is possible for example to introduce a gene into an organism, replace an existing gene with another gene, increase the copy number of the gene(s), use a strong promoter or use a gene which encodes for a corresponding polypeptide with a high activity; optionally, these measures can be combined.

Preferably such constructs according to the invention comprise a promoter 5’- upstream of the respective coding sequence and a terminator sequence 3 ’-downstream and optionally other usual regulatory elements, in each case in operative linkage with the coding sequence.

Nucleic acid constructs according to the invention comprise in particular a sequence coding for a polypeptide for example derived from the amino acid related SEQ ID NOs as described therein or the reverse complement thereof, or derivatives and homologs thereof and which have been linked operatively or functionally with one or more regulatory signals, advantageously for controlling, for example increasing, gene expression.

In addition to these regulatory sequences, the natural regulation of these sequences may still be present before the actual structural genes and optionally may have been genetically modified so that the natural regulation has been switched off and expression of the genes has been enhanced. The nucleic acid construct may, however, also be of simpler construction, i.e. no additional regulatory signals have been inserted before the coding sequence and the natural promoter, with its regulation, has not been removed. Instead, the natural regulatory sequence is mutated such that regulation no longer takes place and the gene expression is increased.

A preferred nucleic acid construct advantageously also comprises one or more of the already mentioned “enhancer” sequences in functional linkage with the promoter, which sequences make possible an enhanced expression of the nucleic acid sequence. Additional advantageous sequences may also be inserted at the 3 ’-end of the DNA sequences, such as further regulatory elements or terminators. One or more copies of the nucleic acids according to the invention may be present in a construct. In the construct, other markers, such as genes which complement auxotrophisms or antibiotic resistances, may also optionally be present so as to select for the construct.

Examples of suitable regulatory sequences are present in promoters such as cos, tac, trp, tet, trp-tet, lpp, lac, lpp-lac, laclq, T7, T5, T3, gal, trc, ara, rhaP (rhaPBAD)SP6, lambda- PR or in the lambda-PL promoter, and these are advantageously employed in Gram-negative bacteria. Further advantageous regulatory sequences are present for example in the Gram positive promoters amy and SP02, in the yeast or fungal promoters ADC1, MFalpha, AC, P- 60, CYC1, GAPDH, TEF, rp28, ADH. Artificial promoters may also be used for regulation.

For expression in a host cell or host organism, the nucleic acid construct is inserted advantageously into a vector such as, for example, a plasmid or a phage, which makes possible optimal expression of the genes in the host. Vectors are also understood as meaning, in addition to plasmids and phages, all the other vectors which are known to the skilled worker, that is to say for example viruses such as SV40, CMV, baculovirus and adenovirus, transposons, IS elements, phasmids, cosmids and linear or circular DNA or artificial chromosomes. These vectors are capable of replicating autonomously in the host organism or else chromosomally. These vectors are a further development of the invention. Binary or co integration vectors are also applicable.

Suitable plasmids are, for example, in E. coll pFG338, pACYC184, pBR322, pUC18, pUC19, pKC30, pRep4, pHSl, pKK223-3, pDHE19.2, pHS2, pPFc236, pMBF24, pFG200, pUR290, pIN-III113-Bl, gtll or pBdCI, in Streptomyces pIJlOl, pIJ364, pIJ702 or pIJ361, in Bacillus pUBllO, pC194 or pBD214, in Corynebacterium pSA77 or pAJ667, in fungi pAFSl, pIF2 or pBB116, in yeasts 2alphaM, pAG-1, YEp6, YEpl3 or pEMBFYe23 or in plants pFGV23, pGHlac+, pBIN19, pAK2004 or pDH5F The abovementioned plasmids are a small selection of the plasmids which are possible. Further plasmids are well known to the skilled worker and can be found for example in the book Cloning Vectors (Eds. Pouwels P. H. et al. Elsevier, Amsterdam-New York-Oxford, 1985, ISBN 0444 904018).

In a further development of the vector, the vector which comprises the nucleic acid construct according to the invention or the nucleic acid according to the invention can advantageously also be introduced into the microorganisms in the form of a linear DNA and integrated into the host cell’s or host organism’s genome via heterologous or homologous recombination. This linear DNA can consist of a linearized vector such as a plasmid or only of the nucleic acid construct or the nucleic acid according to the invention.

For optimal expression of heterologous genes in organisms, it is advantageous to modify the nucleic acid sequences to match the specific “codon usage” used in the organism. The “codon usage” can be determined readily by computer evaluations of other, known genes of the organism in question.

An expression cassette according to the invention is generated by fusing a suitable promoter to a suitable coding nucleotide sequence and a terminator or polyadenylation signal. Customary recombination and cloning techniques are used for this purpose, as are described, for example, in T. Maniatis, E.F. Fritsch and J. Sambrook, Molecular Cloning: A Faboratory Manual, Cold Spring Harbor Faboratory, Cold Spring Harbor, NY (1989) and in T.J. Silhavy, M.F. Berman and F.W. Enquist, Experiments with Gene Fusions, Cold Spring Harbor Faboratory, Cold Spring Harbor, NY (1984) and in Ausubel, F.M. el al., Current Protocols in Molecular Biology, Greene Publishing Assoc and Wiley Interscience (1987).

For expression in a suitable host cell or host organism, the recombinant nucleic acid construct or gene construct is advantageously inserted into a host-specific vector which makes possible optimal expression of the genes in the host. Vectors are well known to the skilled worker and can be found for example in “cloning vectors” (Pouwels P. H. et al., Ed., Elsevier, Amsterdam-New York-Oxford, 1985).

An alternative embodiment of an embodiment herein provides a method to “alter (modify) gene expression” in a host cell. For instance, the polynucleotide of an embodiment herein may be enhanced or overexpressed or induced in certain contexts (e.g. upon exposure to certain temperatures or culture conditions) in a host cell or host organism.

Alteration of expression of a polynucleotide provided herein may also result in ectopic expression which is a different expression pattern in an altered and in a control or wild-type organism. Alteration of expression occurs from interactions of the polypeptide of an embodiment herein with exogenous or endogenous modulators, or as a result of chemical modification of the polypeptide. The term also refers to an altered expression pattern of the polynucleotide of an embodiment herein which is altered below the detection level or completely suppressed activity.

In one embodiment, provided herein is also an isolated, recombinant or synthetic polynucleotide encoding a polypeptide or variant polypeptide provided herein.

In one embodiment, several polypeptide encoding nucleic acid sequences are co expressed in a single host, particularly under control of different promoters. In another embodiment, several polypeptide encoding nucleic acid sequences can be present on a single transformation vector or be co-transformed at the same time using separate vectors and selecting transformants comprising both chimeric genes. Similarly, one or more polypeptide encoding genes may be expressed in a single plant, cell, microorganism or organism together with other chimeric genes. g. Hosts to be applied for the present invention

Depending on the context, the term “host” can mean the wild-type host or a genetically modified (altered), “recombinant host” or both.

In principle, all prokaryotic or eukaryotic organisms may be considered as host or recombinant host organisms for the nucleic acids or the nucleic acid constructs according to the invention.

Using the vectors according to the invention, recombinant hosts can be produced, which are for example transformed with at least one vector according to the invention and can be used for producing the polypeptides according to the invention. Advantageously, the recombinant constructs according to the invention, described above, are introduced into a suitable host system and expressed. Preferably common cloning and transfection methods, known by a person skilled in the art, are used, for example co-precipitation, protoplast fusion, electroporation, retroviral transfection and the like, for expressing the stated nucleic acids in the respective expression system. Suitable systems are described for example in Current Protocols in Molecular Biology, F. Ausubel el ah, Ed., Wiley Interscience, New York 1997, or Sambrook et al. Molecular Cloning: A Laboratory Manual. 2 nd edition, Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989.

Advantageously, microorganisms such as bacteria, fungi or yeasts are used as host organisms. Advantageously, Gram-positive or Gram-negative bacteria are used, preferably bacteria of the families Enterobacteriaceae, Pseudomonadaceae, Rhizobiaceae, Streptomycetaceae, Streptococcaceae or Nocardiaceae, especially preferably bacteria of the genera Escherichia, Pseudomonas, Streptomyces, Lactococcus, Nocardia, Burkholderia, Salmonella, Agrobacterium, Clostridium or Rhodococcus. The genus and species Escherichia coli is quite especially preferred. Furthermore, other advantageous bacteria are to be found in the group of alpha-Proteobacteria, beta-Proteobacteria or gamma-Proteobacteria. Advantageously also yeasts of families like Saccharomyces or Pichia are suitable hosts.

Alternatively, entire plants or plant cells may serve as natural or recombinant host. As non-limiting examples the following plants or cells derived therefrom may be mentioned: the genera Nicotiana, in particular Nicotiana benthamiana and Nicotiana tabacum (tobacco); as well as Arabidopsis, in particular Arabidopsis thaliana. Depending on the host organism, the organisms used in the method according to the invention are grown or cultured in a manner known by a person skilled in the art. Culture can be batchwise, semi-batchwise or continuous. Nutrients can be present at the beginning of fermentation or can be supplied later, semi-continuously or continuously. This is also described in more detail below. h. Recombinant production of polypeptides according to the invention

The invention further relates to methods for recombinant production of polypeptides according to the invention or functional, biologically active fragments thereof, wherein a polypeptide-producing microorganism is cultured, optionally the expression of the polypeptides is induced by applying at least one inducer inducing gene expression and the expressed polypeptides are isolated from the culture. The polypeptides can also be produced in this way on an industrial scale, if desired.

The microorganisms produced according to the invention can be cultured continuously or discontinuously in the batch method or in the fed-batch method or repeated fed-batch method. A summary of known cultivation methods can be found in the textbook by Chmiel (Bioprozesstechnik 1. Einfiihrung in die Bioverfahrenstechnik [Bioprocess technology 1. Introduction to bioprocess technology] (Gustav Fischer Verlag, Stuttgart, 1991)) or in the textbook by Storhas (Bioreaktoren und periphere Einrichtungen [Bioreactors and peripheral equipment] (Vieweg Verlag, Braunschweig/Wiesbaden, 1994)).

The culture medium to be used must suitably meet the requirements of the respective strains. Descriptions of culture media for various microorganisms are given in the manual "Manual of Methods for General Bacteriology" of the American Society for Bacteriology (Washington D. C, USA, 1981).

These media usable according to the invention usually comprise one or more carbon sources, nitrogen sources, inorganic salts, vitamins and/or trace elements.

Preferred carbon sources are sugars, such as mono-, di- or polysaccharides. Very good carbon sources are for example glucose, fructose, mannose, galactose, ribose, sorbose, ribulose, lactose, maltose, sucrose, raffinose, starch or cellulose. Sugars can also be added to the media via complex compounds, such as molasses, or other by-products of sugar refining. It can also be advantageous to add mixtures of different carbon sources. Other possible carbon sources are oils and fats, for example soybean oil, sunflower oil, peanut oil and coconut oil, fatty acids, for example palmitic acid, stearic acid or linoleic acid, alcohols, for example glycerol, methanol or ethanol and organic acids, for example acetic acid or lactic acid. Nitrogen sources are usually organic or inorganic nitrogen compounds or materials that contain these compounds. Examples of nitrogen sources comprise ammonia gas or ammonium salts, such as ammonium sulfate, ammonium chloride, ammonium phosphate, ammonium carbonate or ammonium nitrate, nitrates, urea, amino acids or complex nitrogen sources, such as com- steep liquor, soya flour, soya protein, yeast extract, meat extract and others. The nitrogen sources can be used alone or as a mixture.

Inorganic salt compounds that can be present in the media comprise the chloride, phosphorus or sulfate salts of calcium, magnesium, sodium, cobalt, molybdenum, potassium, manganese, zinc, copper and iron.

Inorganic sulfur-containing compounds, for example sulfates, sulfites, dithionites, tetrathionates, thiosulfates, sulfides, as well as organic sulfur compounds, such as mercaptans and thiols, can be used as the sulfur source.

Phosphoric acid, potassium dihydrogen phosphate or dipotassium hydrogen phosphate or the corresponding sodium-containing salts can be used as the phosphorus source.

Chelating agents can be added to the medium, in order to keep the metal ions in solution. Especially suitable chelating agents comprise dihydroxyphenols, such as catechol or protocatechuate, or organic acids, such as citric acid.

The fermentation media used according to the invention usually also contain other growth factors, such as vitamins or growth promoters, which include for example biotin, riboflavin, thiamine, folic acid, nicotinic acid, pantothenate and pyridoxine. Growth factors and salts often originate from the components of complex media, such as yeast extract, molasses, com-steep liquor and the like. Moreover, suitable precursors can be added to the culture medium. The exact composition of the compounds in the medium is strongly dependent on the respective experiment and is decided for each specific case individually. Information on media optimization can be found in the textbook “Applied Microbiol. Physiology, A Practical Approach" (Ed. P.M. Rhodes, P.F. Stanbury, IRL Press (1997) p. 53- 73, ISBN 0 19963577 3). Growth media can also be obtained from commercial suppliers, such as Standard 1 (Merck) or BHI (brain heart infusion, DIFCO) and the like.

All components of the medium are sterilized, either by heat (20 min at 1.5 bar and 121°C) or by sterile filtration. The components can either be sterilized together, or separately if necessary. All components of the medium can be present at the start of culture or can be added either continuously or batchwise.

The culture temperature is normally between 15°C and 45 °C, preferably 25 °C to 40°C and can be varied or kept constant during the experiment. The pH of the medium should be in the range from 5 to 8.5, preferably around 7.0. The pH for growing can be controlled during growing by adding basic compounds such as sodium hydroxide, potassium hydroxide, ammonia or ammonia water or acid compounds such as phosphoric acid or sulfuric acid. Antifoaming agents, for example fatty acid polyglycol esters, can be used for controlling foaming. To maintain the stability of plasmids, suitable selective substances, for example antibiotics, can be added to the medium. To maintain aerobic conditions, oxygen or oxygen- containing gas mixtures, for example ambient air, are fed into the culture. The temperature of the culture is normally in the range from 20°C to 45 °C. The culture is continued until a maximum of the desired product has formed. This target is normally reached within 10 hours to 160 hours.

The fermentation broth is then processed further. Depending on requirements, the biomass can be removed from the fermentation broth completely or partially by separation techniques, for example centrifugation, filtration, decanting or a combination of these methods or can be left in it completely.

If the polypeptides are not secreted in the culture medium, the cells can also be lysed and the product can be obtained from the lysate by known methods for isolation of proteins. The cells can optionally be disrupted with high-frequency ultrasound, high pressure, for example in a French press, by osmolysis, by the action of detergents, lytic enzymes or organic solvents, by means of homogenizers or by a combination of several of the aforementioned methods.

The polypeptides can be purified by known chromatographic techniques, such as molecular sieve chromatography (gel filtration), such as Q-sepharose chromatography, ion exchange chromatography and hydrophobic chromatography, and with other usual techniques such as ultrafiltration, crystallization, salting-out, dialysis and native gel electrophoresis. Suitable methods are described for example in Cooper, T. G., Biochemische Arbeitsmethoden [Biochemical processes], Verlag Walter de Gruyter, Berlin, New York or in Scopes, R., Protein Purification, Springer Verlag, New York, Heidelberg, Berlin.

For isolating the recombinant protein, it can be advantageous to use vector systems or oligonucleotides, which lengthen the cDNA by defined nucleotide sequences and therefore code for altered polypeptides or fusion proteins, which for example serve for easier purification. Suitable modifications of this type are for example so-called “tags” functioning as anchors, for example the modification known as hexa-histidine anchor or epitopes that can be recognized as antigens of antibodies (described for example in Harlow, E. and Lane, D., 1988, Antibodies: A Laboratory Manual. Cold Spring Harbor (N.Y.) Press). These anchors can serve for attaching the proteins to a solid carrier, for example a polymer matrix, which can for example be used as packing in a chromatography column, or can be used on a microtiter plate or on some other carrier.

At the same time these anchors can also be used for recognition of the proteins. For recognition of the proteins, it is moreover also possible to use usual markers, such as fluorescent dyes, enzyme markers, which form a detectable reaction product after reaction with a substrate, or radioactive markers, alone or in combination with the anchors for derivatization of the proteins. i. Polypeptide immobilization

The enzymes or polypeptides according to the invention can be used free or immobilized in the method described herein. An immobilized enzyme is an enzyme that is fixed to an inert carrier. Suitable carrier materials and the enzymes immobilized thereon are known from EP-A-1149849, EP-A-1 069 183 and DE-OS 100193773 and from the references cited therein. Reference is made in this respect to the disclosure of these documents in their entirety. Suitable carrier materials include for example clays, clay minerals, such as kaolinite, diatomaceous earth, perlite, silica, aluminum oxide, sodium carbonate, calcium carbonate, cellulose powder, anion exchanger materials, synthetic polymers, such as polystyrene, acrylic resins, phenol formaldehyde resins, polyurethanes and polyolefins, such as polyethylene and polypropylene. For making the supported enzymes, the carrier materials are usually employed in a finely-divided, particulate form, porous forms being preferred. The particle size of the carrier material is usually not more than 5 mm, in particular not more than 2 mm (particle-size distribution curve). Similarly, when using dehydrogenase as whole-cell catalyst, a free or immobilized form can be selected. Carrier materials are e.g. Ca-alginate, and carrageenan. Enzymes as well as cells can also be crosslinked directly with glutaraldehyde (cross-linking to CLEAs). Corresponding and other immobilization techniques are described for example in J. Lalonde and A. Margolin immobilization of Enzymes" in K. Drauz and H. Waldmann, Enzyme Catalysis in Organic Synthesis 2002, Vol. Ill, 991-1032, Wiley-VCH, Weinheim. Further information on biotransformations and bioreactors for carrying out methods according to the invention are also given for example in Rehm et al. (Ed.) Biotechnology, 2 nd Edn, Vol 3, Chapter 17, VCH, Weinheim. j. Reaction conditions for biocatalytic production methods of the invention An “enzymatically catalyzed" or “biocatalytic” method means that said method is performed under the catalytic action of an enzyme, including enzyme mutants, as herein defined. Thus, the method can either be performed in the presence of said enzyme in isolated (purified, enriched) or crude form or in the presence of a cellular system, in particular, natural or recombinant microbial cells containing said enzyme in active form, and having the ability to catalyze the conversion reaction as disclosed herein. Hence, the reaction of the present invention may be performed under in vivo or in vitro conditions.

The at least one polypeptide/enzyme which is present during a method of the invention or an individual step of a multistep-method as defined herein above, can be present in living cells naturally or recombinantly producing the enzyme or enzymes, in harvested cells i.e. under in vivo conditions (bioconversion), or, in dead cells, in permeabilized cells, in crude cell extracts, in purified extracts, or in essentially pure or completely pure form, i.e. under in vitro conditions (biochemical synthesis). The at least one enzyme may be present in solution or as an enzyme immobilized on a carrier. One or several enzymes may simultaneously be present in soluble and/or immobilized form.

The methods according to the invention can be performed in common reactors, which are known to those skilled in the art, and in different ranges of scale, e.g. from a laboratory scale (few millilitres to dozens of litres of reaction volume) to an industrial scale (several litres to thousands of cubic meters of reaction volume). If the polypeptide is used in a form encapsulated by non-living, optionally permeabilized cells, in the form of a more or less purified cell extract or in purified form, a chemical reactor can be used. The chemical reactor usually allows controlling the amount of the at least one enzyme, the amount of the at least one substrate, the pH, the temperature and the circulation of the reaction medium. When the at least one polypeptide/enzyme is present in living cells, the process will be a fermentation. In this case the biocatalytic production will take place in a bioreactor (fermenter), where parameters necessary for suitable living conditions for the living cells (e.g. culture medium with nutrients, temperature, aeration, presence or absence of oxygen or other gases, antibiotics, and the like) can be controlled. Those skilled in the art are familiar with chemical reactors or bioreactors, e.g. with procedures for up-scaling chemical or biotechnological methods from laboratory scale to industrial scale, or for optimizing process parameters, which are also extensively described in the literature (for biotechnological methods see e.g. Crueger und Crueger, Biotechnologie - Lehrbuch der angewandten Mikrobiologie, 2. Ed., R. Oldenbourg Verlag, Miinchen, Wien, 1984). Cells containing the at least one enzyme can be permeabilized by physical or mechanical means, such as ultrasound or radiofrequency pulses, French presses, or chemical means, such as hypotonic media, lytic enzymes and detergents present in the medium, or combination of such methods. Examples for detergents are digitonin, n-dodecylmaltoside, octylglycoside, Triton® X-100, Tween ® 20, deoxycholate, CHAPS (3-[(3-

Cholamidopropyl)dimethylammonio]-l -propansulfonate), Nonidet ® P40

(Ethylphenolpoly(ethyleneglycolether), and the like.

Instead of living cells biomass of non-living cells containing the required biocatalyst(s) may be applied of the biotransformation reactions of the invention as well.

If the at least one enzyme is immobilized, it is attached to an inert carrier as described above.

The conversion reaction can be carried out batch wise, semi-batch wise or continuously. Reactants (and optionally nutrients) can be supplied at the start of reaction or can be supplied subsequently, either semi-continuously or continuously.

The reaction of the invention, depending on the particular reaction type, may be performed in an aqueous, aqueous-organic or non-aqueous reaction medium.

An aqueous or aqueous-organic medium may contain a suitable buffer in order to adjust the pH to a value in the range of 5 to 11, like 6 to 10.

In an aqueous-organic medium an organic solvent miscible, partly miscible or immiscible with water may be applied. Non-limiting examples of suitable organic solvents are listed below. Further examples are mono- or polyhydric, aromatic or aliphatic alcohols, in particular polyhydric aliphatic alcohols like glycerol.

The non-aqueous medium may be substantially free of water, i.e. will contain less that about 1 wt.-% or 0.5 wt.-% of water.

Biocatalytic methods may also be performed in an organic non-aqueous medium. As suitable organic solvents there may be mentioned aliphatic hydrocarbons having for example 5 to 8 carbon atoms, like pentane, cyclopentane, hexane, cyclohexane, heptane, octane or cyclooctane; aromatic carbohydrates, like benzene, toluene, xylenes, chlorobenzene or dichlorobenzene, aliphatic acyclic and ethers, like diethylether, methyl-tert.-butylether, ethyl- tert.-butylether, dipropylether, diisopropylether, dibutylether; or mixtures thereof.

The concentration of the reactants/substrates may be adapted to the optimum reaction conditions, which may depend on the specific enzyme applied. For example, the initial substrate concentration may be in the 0.1 to 0.5 M, as for example 10 to 100 mM. The reaction temperature may be adapted to the optimum reaction conditions which may depend on the specific enzyme applied. For example, the reaction may be performed at a temperature in a range of from 0 to 70°C, as for example 20 to 50 or 25 to 40°C. Examples for reaction temperatures are about 30°C, about 35°C, about 37°C, about 40°C, about 45°C, about 50°C, about 55°C and about 60°C.

The process may proceed until equilibrium between the substrate and then product(s) is achieved, but may be stopped earlier. Usual process times are in the range from 1 minute to 25 hours, in particular 10 min to 6 hours, as for example in the range from 1 hour to 4 hours, in particular 1.5 hours to 3.5 hours. These parameters are non-limiting examples of suitable process conditions.

If the host is a transgenic plant, optimal growth conditions can be provided, such as optimal light, water and nutrient conditions, for example.

1. Product isolation

The methodology of the present invention can further include a step of recovering an end or intermediate product, optionally in stereoisomerically or enantiomerically substantially pure form. The term “recovering” includes extracting, harvesting, isolating or purifying the compound from culture or reaction media. Recovering the compound can be performed according to any conventional isolation or purification methodology known in the art including, but not limited to, treatment with a conventional resin (e.g., anion or cation exchange resin, non-ionic adsorption resin, etc.), treatment with a conventional adsorbent (e.g., activated charcoal, silicic acid, silica gel, cellulose, alumina, etc.), alteration of pH, solvent extraction (e.g., with a conventional solvent such as an alcohol, ethyl acetate, hexane and the like), distillation, dialysis, filtration, concentration, crystallization, recrystallization, pH adjustment, lyophilization and the like.

Identity and purity of the isolated product may be determined by known techniques, like High Performance Liquid Chromatography (HPLC), Gas Chromatography (GC), Spectroscopy (like IR, UV, NMR), Colouring methods, TLC, NIRS, enzymatic or microbial assays (see for example: Patek et al. (1994) Appl. Environ. Microbiol. 60:133-140; Malakhova et al. (1996) Biotekhnologiya 11 27-32; and Schmidt et al. (1998) Bioprocess Engineer. 19:67-70. Ullmann's Encyclopedia of Industrial Chemistry (1996) Bd. A27, VCH: Weinheim, p. 89-90, p. 521-540, p. 540-547, p. 559-566, 575-581 and p. 581-587; Michal, G (1999) Biochemical Pathways: An Atlas of Biochemistry and Molecular Biology, John Wiley and Sons; Fallon, A. et al. (1987) Applications of HPLC in Biochemistry in: Laboratory Techniques in Biochemistry and Molecular Biology, Bd. 17).

The cyclic terpene compound produced in any of the methods described herein can be converted to derivatives such as, but not limited to hydrocarbons, esters, amides, glycosides, ethers, epoxides, aldehydes, cycloaddition products, ketones, alcohols, diols, acetals or ketals. The terpene compound derivatives can be obtained by a chemical method such as, but not limited to oxidation, reduction, alkylation, acylation and/or rearrangement. Alternatively, the terpene compound derivatives can be obtained using a biochemical method by contacting the terpene compound with an enzyme such as, but not limited to an oxidoreductase, a monooxygenase, esterase, a dioxygenase, a transferase. The biochemical conversion can be performed in vitro using isolated enzymes, enzymes from lysed cells or in vivo using whole cells. m. Fermentative production of drimenyl diphosphate and/or drimenol, and/or albicanyl diphosphate and/or albicanol

The invention also relates to methods for the fermentative production of drimenyl diphosphate and/or drimenol, and/or albicanyl diphosphate and/or albicanol.

The term “fermentative production” or “fermentation” refers to the ability of a microorganism (assisted by enzyme activity contained in or generated by said microorganism) to produce a chemical compound in cell culture utilizing at least one carbon source added to the incubation.

The term “fermentation broth“ is understood to mean a liquid, particularly aqueous or aqueous/organic solution which is based on a fermentative process and has not been worked up or has been worked up, for example, as described herein.

A fermentation as used according to the present invention can, for example, be performed in stirred fermenters, bubble columns and loop reactors. A comprehensive overview of the possible method types including stirrer types and geometric designs can be found in “Chmiel: Bioprozesstechnik: Einfiihrung in die Bioverfahrenstechnik, Band 1 [Bioprocess technology 1. Introduction to bioprocess technology]”. In the process of the invention, typical variants available are the following variants known to those skilled in the art or explained, for example, in “Chmiel, Hammes and Bailey: Biochemical Engineering", such as batch, fed-batch, repeated fed-batch or else continuous fermentation with and without recycling of the biomass. Depending on the production strain, sparging with air, oxygen, carbon dioxide, hydrogen, nitrogen or appropriate gas mixtures may be effected in order to achieve good yield (Yp/S).

“Yield" and/or the “conversion rate” of a reaction according to the invention is determined over a defined period of, for example, 4, 6, 8, 10, 12, 16, 20, 24, 36 or 48 hours, in which the reaction takes place. In particular, the reaction is carried out under precisely defined conditions, for example at “standard conditions” as herein defined.

The different yield parameters (“Yield” or Yp / s; “Specific Productivity Yield”; or Space-Time-Yield (STY)) are well known in the art and are determined as described in the literature.

“Yield" and “Yp / s” (each expressed in mass of product produced/mass of material consumed) are herein used as synonyms.

The specific productivity-yield describes the amount of a product that is produced per h and L fermentation broth per g of biomass. The amount of wet cell weight stated as WCW describes the quantity of biologically active microorganism in a biochemical reaction. The value is given as g product per g WCW per h (i.e. g gwcw 1 h 1 ). Alternatively, the quantity of biomass can also be expressed as the amount of dry cell weight stated as DCW. Furthermore, the biomass concentration can be more easily determined by measuring the optical density at 600 nm (ODeoo) and by using an experimentally determined correlation factor for estimating the corresponding wet cell or dry cell weight, respectively.

The culture medium that is to be used according to the present invention must satisfy the requirements of the particular strains in an appropriate manner. Descriptions of culture media for various microorganisms are given in the handbook “Manual of Methods for General Bacteriology” of the American Society for Bacteriology (Washington D. C., USA, 1981).

These media that can be used according to the invention may comprise one or more sources of carbon, sources of nitrogen, inorganic salts, vitamins and/or trace elements.

Preferred sources of carbon are sugars, such as mono-, di- or polysaccharides. Very good sources of carbon are for example glucose, fructose, mannose, galactose, ribose, sorbose, ribulose, lactose, maltose, sucrose, raffinose, starch or cellulose. Sugars can also be added to the media via complex compounds, such as molasses, or other by-products from sugar refining. It may also be advantageous to add mixtures of various sources of carbon. Other possible sources of carbon are oils and fats such as soybean oil, sunflower oil, peanut oil and coconut oil, fatty acids such as palmitic acid, stearic acid or linoleic acid, alcohols such as glycerol, methanol or ethanol and organic acids such as acetic acid or lactic acid. Sources of nitrogen are usually organic or inorganic nitrogen compounds or materials containing these compounds. Examples of sources of nitrogen include ammonia gas or ammonium salts, such as ammonium sulfate, ammonium chloride, ammonium phosphate, ammonium carbonate or ammonium nitrate, nitrates, urea, amino acids or complex sources of nitrogen, such as corn- steep liquor, soybean flour, soy-bean protein, yeast extract, meat extract and others. The sources of nitrogen can be used separately or as a mixture.

Inorganic salt compounds that may be present in the media comprise the chloride, phosphate or sulfate salts of calcium, magnesium, sodium, cobalt, molybdenum, potassium, manganese, zinc, copper and iron.

Inorganic sulfur-containing compounds, for example sulfates, sulfites, di-thionites, tetrathionates, thiosulfates, sulfides, but also organic sulfur compounds, such as mercaptans and thiols, can be used as sources of sulfur.

Phosphoric acid, potassium dihydrogenphosphate or dipotassium hydrogenphosphate or the corresponding sodium-containing salts can be used as sources of phosphorus.

Chelating agents can be added to the medium, in order to keep the metal ions in solution. Especially suitable chelating agents comprise dihydroxyphenols, such as catechol or protocatechuate, or organic acids, such as citric acid.

The fermentation media used according to the invention may also contain other growth factors, such as vitamins or growth promoters, which include for example biotin, riboflavin, thiamine, folic acid, nicotinic acid, pantothenate and pyridoxine. Growth factors and salts often come from complex components of the media, such as yeast extract, molasses, corn- steep liquor and the like. In addition, suitable precursors can be added to the culture medium. The precise composition of the compounds in the medium is strongly dependent on the particular experiment and must be decided individually for each specific case. Information on media optimization can be found in the textbook “Applied Microbiol. Physiology, A Practical Approach" (1997) Growing media can also be obtained from commercial suppliers, such as Standard 1 (Merck) or BHI (Brain heart infusion, DIFCO) etc.

All components of the medium are sterilized, either by heating (20 min at 1.5 bar and 121°C) or by sterile filtration. The components can be sterilized either together, or if necessary separately. All the components of the medium can be present at the start of growing, or optionally can be added continuously or by batch feed.

The temperature of the culture is normally between 15°C and 45°C, preferably 25°C to 40°C and can be kept constant or can be varied during the experiment. The pH value of the medium should be in the range from 5 to 8.5, preferably around 7.0. The pH value for growing can be controlled during growing by adding basic compounds such as sodium hydroxide, potassium hydroxide, ammonia or ammonia water or acid compounds such as phosphoric acid or sulfuric acid. Antifoaming agents, e.g. fatty acid polyglycol esters, can be used for controlling foaming. To maintain the stability of plasmids, suitable substances with selective action, e.g. antibiotics, can be added to the medium. Oxygen or oxygen-containing gas mixtures, e.g. the ambient air, are fed into the culture in order to maintain aerobic conditions. The temperature of the culture is normally from 20°C to 45 °C. Culture is continued until a maximum of the desired product has formed. This is normally achieved within 1 hour to 160 hours.

The methodology of the present invention can further include a step of recovering drimenyl diphosphate and/or drimenol, and/or albicanyl diphosphate and/or albicanol

The term “recovering” includes extracting, harvesting, isolating or purifying the compound from culture media. Recovering the compound can be performed according to any conventional isolation or purification methodology known in the art including, but not limited to, treatment with a conventional resin (e.g., anion or cation exchange resin, non-ionic adsorption resin, etc.), treatment with a conventional adsorbent (e.g., activated charcoal, silicic acid, silica gel, cellulose, alumina, etc.), alteration of pH, solvent extraction (e.g., with a conventional solvent such as an alcohol, ethyl acetate, hexane and the like), distillation, dialysis, filtration, concentration, crystallization, recrystallization, pH adjustment, lyophilization and the like.

Before the intended isolation the biomass of the broth can be removed. Processes for removing the biomass are known to those skilled in the art, for example filtration, sedimentation and flotation. Consequently, the biomass can be removed, for example, with centrifuges, separators, decanters, filters or in flotation apparatus. For maximum recovery of the product of value, washing of the biomass is often advisable, for example in the form of a diafiltration. The selection of the method is dependent upon the biomass content in the fermenter broth and the properties of the biomass, and also the interaction of the biomass with the product of value.

In one embodiment, the fermentation broth can be sterilized or pasteurized. In a further embodiment, the fermentation broth is concentrated. Depending on the requirement, this concentration can be done batch wise or continuously. The pressure and temperature range should be selected such that firstly no product damage occurs, and secondly minimal use of apparatus and energy is necessary. The skillful selection of pressure and temperature levels for a multistage evaporation in particular enables saving of energy. The following examples are illustrative only and are not intended to limit the scope of the claims an embodiments described herein.

The numerous possible variations that will become immediately evident to a person skilled in the art after heaving considered the disclosure provided herein also fall within the scope of the invention.

Experimental Part

The invention will now be described in further detail by way of the following Examples.

Materials:

Unless otherwise stated, all chemical and biochemical materials and microorganisms or cells employed herein are commercially available products.

Unless otherwise specified, recombinant proteins are cloned and expressed by standard methods, such as, for example, as described by Sambrook, J., Fritsch, E.F. and Maniatis, T., Molecular Cloning: A Laboratory Manual, 2 nd Edition, Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989.

Methods:

GC/MS

GC/MS analysis was performed on a GC/MS 6890N/5975 (Agilent). Separation was achieved using a DB-1MS column (30 m x 0.25 mm i.d. x 0.25 pm film thickness, J & W Scientific, Agilent Technologies, Foster City, CA). The carrier gas was helium at a constant flow of 0.7 mL/min. Injection was in split (1:5) mode with the injector temperature set at 250°C. The oven temperature was programmed to increase first from 50°C (5 min hold) to 300°C incrementing at 5°C/min, and then to 340°C incrementing at 50°C/min, where it was held for 3 min. Data were acquired by scan mode with m/z 29-450. Alkanes (C6-C30) were injected for calculation of retention index. Compounds were identified by matching mass spectra and retention index with that of in-house library.

Example 1

Discovery of HAD-like TPSs Fresh leaves of Bazzania species (Nagobase ID: PA-2018-0168 to PA-2018-0173, which contain drimane-type sesquiterpene alcohols including albicanol and drimenol) were sourced in Hunan, China. Total RNA was extracted from the fresh leaves using the RNeasy Plant Mini Kit (50) 74904 from QIAGEN and used for RNA sequencing. The RNA sequencing data were then assembled by CLC genomics workbench (7.6.1). The sequence of the fem HAD-like TPS, DfHAD, from prior art (PCT/EP2019/063824; herein SEQ ID NOs: 73 and 74) was used for searching the potential drimane-type sesquiterpene synthase in the protein sequence data of Bazzania species. Novel sequences were discovered from both assemblies of Bazzania transcriptomes by BLAST, resulting in HAD-like TPSs designated BazzHADl (SEQ ID NO: 3), BazzHAD2 (SEQ ID NO: 6) and BazzHAD3 (SEQ ID NO: 9); their corresponding nucleotide sequences are SEQ ID NO: 1, 4 and 7, respectively.

The RNAseq raw data of Bazzania trilobata (SRA ID: ERR364415) was downloaded from NCBI and assembled by CLC genomics workbench to deliver the transcriptome from which the BtHAD (SEQ ID NO: 12) was discovered by BLAST; its corresponding nucleotide sequence is SEQ ID NO: 10.

Homologous sequences of DfHAD in the NCBI database were thoroughly mined by using BLAST with threshold scores (E-value<10, Hit list size<500) until plant genes were observed, resulting in two similar sequences from Selaginella moellendorjfii that were deposited with accession numbers EFJ10816.1 and EFJ26126.1 (GenBank ID) as hypothetical proteins, not relevant to the said HAD-like TPS. The proteins were respectively designated SmHADl (SEQ ID NO: 19) and SmHAD2 (SEQ ID NO: 26) in this study; their corresponding nucleotide sequences are SEQ ID NOs: 17 and 24, respectively.

To study their homology, an alignment was performed by Clustalw for SmHADl, SmHAD2, BtHAD, BazzHADl, BazzHAD2 and BazzHAD3 together with DfHAD (PCT/EP2019/063824) and several fungal HAD-like drimane sesquiterpene synthases including AstC (PCT/EP2019/063824). We found that the protein sequences of SmHADl and SmHAD2 have a very low degree of identity with all other known HAD-like TPSs (-14-20%) but show high homology between themselves (>96%), BtHAD and BazzHADl show a slightly higher homology to fern and fungal HAD-like TPSs (-22-30%) and 96% identity between themselves, whereas BazzHAD2 and BazzHAD3 show an intermediate homology to fem and fungal HAD-like TPSs (-22-43%) and 67% identity between themselves (Table 1). ble 2. Pairwise comparisons of BazzHADl, BazzHAD2, BazzHAD3, BtHAD, SmHADl, SmHAD2 and known HAD-like TPSs. The mparison shows identity of the aligned sequence between proteins.

Example 2

Functional expression and characterization of the plant HAD-like TPSs in an E. coli expression system ( in vivo biochemical assay)

The coding sequences of BazzHADl, BazzHAD2, BazzHAD3, BtHAD, SmHADl (EFJ10816.1) and SmHAD2 (EFJ26126.1) were optimized by Genscript following their genetic codon usage frequency table for E. coli (SEQ ID NOs: 2, 5, 8, 11, 18 and 25, respectively). They were then synthesized in vitro and subcloned into the pETDuet-1 plasmid (Novagen) for subsequent expression in E. coli, generating the plasmids pETDuet- BazzHADl, pETDuet-BazzHAD2, pETDuet-BazzHAD3, pETDuet-BtHAD, pETDuet- SmHADl, and pETDuet-SmHAD2, respectively.

BL21(DE3) E. coli cells (Tiangen) were co-transformed with the plasmid pACYC/ScMVA, containing the genes encoding for a heterologous mevalonate (MVA) pathway, and each of the plasmids pETDuet-BazzHADl, pETDuet-BazzHAD2, pETDuet- BazzHAD3, pETDuet-BtHAD, pETDuet-SmHADl, and pETDuet-SmHAD2, respectively. The pACYC/ScMVA plasmid for the expression of a farnesyl pyrophosphate (FPP) synthase gene and the genes for a complete MVA pathway were constructed. Briefly, the eight biosynthetic genes of the MVA pathway were divided into 2 synthetic operons referred to as the ‘upper’ and ‘lower’ MVA pathway. As an ‘upper’ MVA pathway, a synthetic operon was created consisting of an acetoacetyl-CoA thiolase from E. coli encoded by atoB, a HMG-CoA synthase and a truncated version of HMG-CoA reductase from Saccharomyces cerevisiae encoded by ERG13 and ERG19, respectively. This operon transforms the primary metabolite Acetyl-CoA into (R)-mevalonate. As a ‘lower’ mevalonate pathway, a second synthetic operon was created encoding a mevalonate kinase (ERG12, S. cerevisiae), a phosphomevalonate kinase (ERG8, S. cerevisiae), a phosphomevalonate decarboxylase (MVD1, S. cerevisiae), an isopentenyl diphosphate isomerase (idi, E. coli) and a FPP synthase (IspA, E. coli). Finally, a second FPP synthase from S. cerevisiae (ERG20) was introduced into the upper pathway operon to improve the conversion of the isoprenoid C5 units (IPP and DMAPP) into FPP. Each operon was subcloned into one of the multiple-cloning sites of a low-copy expression plasmid under the control of a bacteriophage T7 promoter (pACYCDuet-1, Invitrogen) providing the plasmid pACYC/ScMVA. This plasmid thus contains the genes encoding all enzymes of the biosynthetic pathway leading from acetyl-coenzyme A to FPP. The co-transformed cells were selected on LB-agar plates containing ampicillin (50 pg/mL) and chloramphenicol (34 pg/mL). Single colonies were used to inoculate 5 mL liquid LB medium supplemented with the same antibiotics. Cultures were incubated overnight at 37°C while shaking at 200 rpm. The next day, 2 mL of TB medium supplemented with the above-mentioned antibiotics and glycerol (3 % w/v final) were inoculated with 0.2 mL of the overnight culture. After 5 hours of incubation at 37°C and shaking at 200 rpm, the culture was cooled down to 25 °C and shaken a further hour at 200 rpm. IPTG (0.1 mM final) was then added to each tube and the culture overlaid with 200 pL of decane. The cultures were incubated for a further 48 hours at 25°C at 200 rpm and then extracted twice with 1 volume of ethyl acetate. 50 pL of isolongifolene at 2 mg/mL were added to the organic phase as internal standard before analysing the samples by GC/MS. The carrier gas was helium at a constant flow of 0.7 mL/min. Injection was in split (1:25) mode with the injector temperature set at 250°C. The oven temperature was programmed to increase first from 50°C (5 min hold) to 300°C incrementing at 5°C/min, and then to 340°C incrementing at 50°C/min where it was held for 3 min. Identification of products was based on mass spectra and retention indices.

GC/MS analysis revealed that recombinant cells expressing BazzHADl and BtHAD produced 37.3 mg/L and 26.0 mg/L drimenol, respectively; the expression of BazzHAD2 resulted in the production of 35.3 mg/L albicanol and 0.2 mg/L unknown sesquiterpene 1 (unknown SQT1); the expression of BazzHAD3 resulted in the production of 422.7 mg/L albicanol; whereas the expression of SmHADl resulted in the production of 0.5 mg/L albicanol and 6.2 mg/L drimenol; and the expression of SmHAD2 resulted in the production of 1.4 mg/L albicanol and 10.7 mg/L drimenol, see Figures 1-16.

Example 3

Importance of the class motif and OW motif in plant HAD-like TPSs (enzyme mutants and functional proofs for the motifs)

To confirm the importance of the class II terpene synthase motif (SEQ ID NO: 46), mutants of this motif were designed and generated for BtHAD, SmHADl and SmHAD2 (SEQ ID NOs: 13, 20 and 27, respectively). As BazzHADl is an ortholog of BtHAD (with 96% identity) from Bazzania trilobata, it can be represented by BtHAD and was thus not included in this study. In addition to the class II motif, the QW motif (SEQ ID NO: 51) was also mutated in BtHAD, SmHADl and SmHAD2 (SEQ ID NOs: 15, 22 and 29, respectively) in order to study its importance for the synthase activity. Since the known fungal HAD-like TPSs contain both class I and class II motifs (e.g., SEQ ID NOs: 56, 57 and 58 of WO 2018/220113), EMD37666.1 (SEQ ID NO: 31) and XP 007369631.1 (SEQ ID NO: 35) were selected and their class II motif mutated (SEQ ID NOs: 33 and 37, respectively) in order to investigate if the class I motif is sufficient for the synthase activity. In the mutants of the class II synthase motif in BtHAD (D320A/D322A), SmHADl (D257A/D259A), SmHAD2 (D257A/D259A), EMD37666.1

(D276 A/S 277 A/D279 A) and XP 007369631.1 (D272A/D273A/D275A), the polar Asp was substituted by Ala (SEQ ID NOs: 39 to 42). In the mutants of the QW motif in BtHAD (D491A), SmHADl (E432A/D433A) and SmHAD2 (E432A/D433A), the polar Glu and Asp were substituted by Ala (SEQ ID NOs: 43 and 44).

The sequences of BtHAD, SmHADl, SmHAD2, XP 007369631.1, EMD37666.1 and their corresponding mutants for the class II synthase motif and - if present - for the QW motif were optimized by Genscript following their genetic codon usage frequency table for E. coli (BtHAD SEQ ID NOs: 11, 13, 15; SmHADl SEQ ID NOs: 18, 20, 22; SmHAD2 SEQ ID NOs: 25, 27, 29; EMD37666.1 SEQ ID NOs: 31, 33; and XP_007369631.1 SEQ ID NOs: 35, 37). The sequences were then synthesized in vitro by Genscript and subcloned into the pETDuet-1 plasmid (Novagen) for subsequent expression in E. coli, generating the plasmids pETDuet-BtHAD, pETDuet-SmHADl, pETDuet- SmHAD2, pETDuet-EMD37666.1, pETDuet-XP_007369631.1; pETDuet-BtHAD-ClassII mut, pETDuet-SmHADl-ClassII mut, pETDuet-SmHAD2-ClassII mut, pETDuet- EMD37666.1-ClassII mut, pETDuet-XP_007369631.1-ClassII mut; and pETDuet-BtHAD- QW mut, pETDuet-SmHADl-QW mut, and pETDuet-SmHAD2-QW mut, respectively.

BL21(DE3) E. coli cells (Tiangen) were co-transformed with the plasmid pACYC/ScMVA, containing the genes encoding for a heterologous mevalonate pathway, and each of the plasmids pETDuet-BtHAD, pETDuet-SmHADl, pETDuet-SmHAD2, pETDuet-EMD37666.1, pETDuet-XP_007369631.1, pETDuet-BtHAD-ClassII mut, pETDuet-SmHADl-ClassII mut, pETDuet-SmHAD2-ClassII mut, pETDuet- EMD37666.1-ClassII mut, pETDuet-XP_007369631.1-ClassII mut, pETDuet-B tHAD-QW mut, pETDuet-SmHADl-QW mut, and pETDuet-SmHAD2- QW mut, respectively. The construction of the pACYC/ScMVA for the expression of an FPP synthase gene and the genes for a complete MVA pathway is described above (Example 2). This plasmid contains the genes encoding all enzymes of the biosynthetic pathway leading from acetyl- coenzyme A to FPP.

The co-transformed cells were selected on LB -agar plates containing ampicillin (50 pg/mL) and chloramphenicol (34 pg/mL). Single colonies were used to inoculate 5 mL liquid LB medium supplemented with the same antibiotics. Cultures were incubated overnight at 37°C while shaking at 200 rpm. The next day, 2 mL of TB medium supplemented with the above-mentioned antibiotics and glycerol (3 % w/v final) were inoculated with 0.2 mL of the overnight culture. After 5 hours of incubation at 37°C and shaking at 200 rpm, the culture was cooled down to 25 °C and shaken a further hour at 200 rpm. IPTG (0.1 mM final) was then added to each tube, and the culture overlaid with 200 pL of decane. The cultures were incubated for a further 48 hours at 25°C at 200 rpm and then extracted twice with 1 volume of ethyl acetate. 50 pL of isolongifolene at 2 mg/mL were added to the organic phase as internal standard before analysing the samples by GC/MS.

BtHAD, SmHADl, SmHAD2, the fungal HAD-like TPSs and their mutants were analysed in an in vivo biochemical assay. In all the enzymes with a mutated class II motif, neither albicanol nor drimenol was detected. These results support the hypothesis of a protonation-initiated cyclisation of FPP, with the involvement of a class II motif, rather than a classic ionisation-initiated cyclisation mediated by a class I motif (Figure 19). We also observed a 90% and higher loss of productivity of albicanol and drimenol by the mutants of QW motif, possibly associated with the enzyme integrity and thermostability.

Example 4

Characterisation of the plant HAD-like TPSs in an in vitro system (proof of the presumably albicanyl and drimenyl diphosphates).

An in vitro biochemical assay was performed to study the cyclisation mechanism of the newly identified plant HAD-like drimane sesquiterpene synthases.

BL21(DE3) E. coli cells (Tiangen) were transformed with the plasmids pETDuet- SmHADl, pETDuet-SmHAD2 and pETDuet-BtHAD, respectively. The transformed cells were selected on LB-agar plates containing ampicillin (50 pg/mL). Single colonies were used to inoculate 25 mL liquid LB medium supplemented with the same antibiotics. The culture was incubated for 5 hours at 37°C at 200 rpm, until an ODeoo of about 0.5 has been reached, was then cooled down to 20°C and shaken a further 0.5 hour at 200 rpm. Then, IPTG (0.1 mM final) was added to each tube and the cultures were incubated for a further 18 hours at 20°C at 200 rpm.

For the in vitro biochemical assay, 25 mL of E. coli culture expressing SmHADl, SmHAD2, or BtHAD, respectively, were centrifuged and re-suspended in 5 mL 50 mM Tris-HCl pH 8.0 buffer (10 mM MgCF, 5 mM DTT), followed by sonication to yield the crude cell lysates. 29 mM FPP were added to 1 mL of each of the crude cell lysates for the reaction and incubated at 30°C, 50 rpm for 2 hours. The samples were then split into two halves and to one of them 20 pL of a bacterial alkaline phosphatase (BAP; Sangon- B004081-100) was added and the reaction pursued for an extra 1 hour at 25°C (BAP can remove the potential diphosphate groups from drimenyl and/or albicanyl diphosphates to yield drimenol and/or albicanol). A control with no BAP (second half of the sample) was incubated under the same conditions. 10 pL of isolongifolene (internal standard) at 2 mg/mL was added into a 0.5 mL aliquot of the organic phase as internal standard. The reactions were then extracted with 0.25 mL ethyl acetate and analysed by GC/MS, the method as described above.

The addition of BAP to the reactions using SmHAD2 and BtHAD lysates resulted in a 3.5-fold and 1.8-fold increase in the amount of drimenol, respectively. The presence of drimenol in the reactions in lack of BAP treatment can be explained by the activity of E. coifs native phosphatases. The reaction using SmHADl lysate did not produce an observable amount of drimenol, but the addition of BAP allowed its detection, albeit in a minute amount. Likewise, only a minute amount of albicanol could be detected in the reaction using SmHAD2 lysate and in the reaction where BAP was added (Figure 20 and Table 3).

Table 3. Productivity of SmHADl and SmHAD2 in in vitro biochemical assay

Identification Titre (mg/L)

These findings support the assumption of protonation-initiated cyclisation mechanism for the biosynthesis of drimenyl and albicanyl diphosphates.

All the publications mentioned in this application are incorporated by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.

Listing of Sequences

Table 4. Sequences described and used herein

SEQ ID NO: 1

Bazzania sp._BazzHADl_wt_nucleic acid sequence

ATGGCTCCTCCTTTCGGATTTATTGGTGGGAATGCAACGAGTGTGACGACTTCCCCA GATGTCCTCGACTACA ACTATGACGGTGGCCCTCCTACTGGCCAATACAAGAGTTTGATTTTAGACATTGGTGGAG TGTTGCTTCAGCC CAACCTTGACAGGAGCTATGGCACGATTGTACCACCTGTGCAATTCAAACGTATGTTGCG CTCGCACATCTGG TTCAATTACTCAATGGGAAAGATGTCTGCGGAGGAAGTGTTTCGGCAGCTCTCCGTGAAG TCTGGCTACCCGG CAGAAGATATCCGGAGTTTTGGTCGCAAAGCGAGAGAGTCTCTTGTCCCCATCACACAAA TAACGGATCTTCT GTTCAAACTCAGCAAGGAATCGAAGGTCAGACTGTTCTGTATGACCAATTGCCCTGCAGA AGACTTCGAATAC CTCTCACAGGCATATCCCAAGCTTTTCGGCTTGTTCGAGAGGATTTTTACGTCGGCATCC ACCGGTATGCGCA AGCCAAACAGGAATTTCTTCCGACATGTACTCCAGGAGACGGGAATCAGAGCGTCGGAGA CTATTTTCGTAGA CGACCTGGTACCGAACATTATGGCCGCCGAGGCGCTGGACTTCACGGCGATTCACGTTTC TCCAGAGTCGCCC GCCGAGACCGTGAGGTTGCTGATGTTCCATCTCCGTAAGCCAGAAGAGCGGCTGGCGGCG GCGCGCGACTATC TCCTGCGCAACCGTGGAGTGGACAAAGCTCTCTCTCTCACATCTGATGGAGTCCGAGTCC AGGTTTATTTCGA CCACTTCGTGGTCGCCGAAGTGCTCAACGACGAGCGATTCTTGCCAATCACTGCAGCTCC GGAGTCCGGGACT ATGAACTTTTTCCCGAAGGAGGAACGCAAAGCGACTCGAGATATAACGACCAATGCAGTC ACACACGTGGACC TTCCCGATGATCTTGACTCCACCTCTGTCGCCTTATCGGTCCTTTACAAATTCTCCAAAG TGGGGATGGAGAC GGTTCAAAGGGTCCTGGATATGATGGAGAAACGTGTGGATGATGACGGCATTTTTCAAAC GTACTTCGATCCT TTGCGGCTGCGTGTTGACCCGGTGGCCGCTACTAACACCGTCTACCTCTTCCATCTAGGA GGTCGTCCCGGAC CTACTCAGCAGACGGAACAATTCCTCTCCAAGTTACTGGAAAAGAGATCTTATCTCCGCG GCACACTCTACTA CCGGACCCCCGAGGCATTTCTCTATCAACTCACCAGATTGGTGGTATCATTTGAGGAGTA TTTCAGAAAGACA GGCTTCCTGGAAATGCTCAAGAAAAGGCTGAGTGAGCGAGTGGGCATAGAGAGCGATGCA TTCACGTTGGCGA TGAGGATTCTAGCGTGTGTGAATTGTGGAATCCCTGTTGCTGAGCTGAACAGAGATATGG ACAGACTAGCACG GATGCAAAATGTGGATGGTTCATGGGACGTCTGCCCTTACTACAGCTACAATGATCCCAA GAGTTGGTTTGGG AACGAGCTTCTTACCACAGCTTTTGCTGCTGCCGCTCTTGAGCAGTCTGAGCCCAACTTT AACTGA

SEQ ID NO: 2

Bazzania sp._BazzHADl_E. coll optimized

ATGGCTCCGCCGTTCGGCTTTATCGGTGGCAACGCGACCAGCGTTACCACCAGCCCG GATGTGCTGGACTACA ACTATGATGGTGGCCCGCCGACCGGCCAGTACAAAAGCCTGATCCTGGATATTGGTGGCG TTCTGCTGCAACC GAACCTGGACCGTAGCTATGGCACCATCGTTCCGCCGGTGCAGTTCAAGCGTATGCTGCG TAGCCACATTTGG TTTAACTACAGCATGGGTAAAATGAGCGCGGAGGAAGTTTTCCGTCAACTGAGCGTGAAG AGCGGTTATCCGG CGGAGGATATCCGTAGCTTTGGCCGTAAAGCGCGTGAAAGCCTGGTTCCGATCACCCAGA TTACCGACCTGCT GTTCAAGCTGAGCAAAGAGAGCAAAGTGCGTCTGTTCTGCATGACCAACTGCCCGGCGGA GGACTTTGAATAC CTGAGCCAAGCGTATCCGAAACTGTTCGGTCTGTTTGAACGTATTTTTACCAGCGCGAGC ACCGGCATGCGTA AGCCGAACCGTAACTTCTTTCGTCACGTGCTGCAGGAGACCGGCATCCGTGCGAGCGAAA CCATTTTCGTTGA CGATCTGGTGCCGAACATCATGGCGGCGGAGGCGCTGGATTTTACCGCGATTCACGTTAG CCCGGAGAGCCCG GCGGAAACCGTGCGTCTGCTGATGTTCCACCTGCGTAAACCGGAGGAACGTCTGGCGGCG GCGCGTGACTACC TGCTGCGTAACCGTGGTGTTGATAAAGCGCTGAGCCTGACCAGCGATGGTGTGCGTGTTC AGGTGTATTTCGA TCACTTTGTGGTTGCGGAAGTGCTGAACGACGAACGTTTTCTGCCGATCACCGCGGCGCC GGAAAGCGGCACC ATGAACTTCTTTCCGAAAGAGGAACGCAAGGCGACCCGTGATATTACCACCAACGCGGTT ACCCACGTGGATC TGCCGGACGATCTGGACAGCACCAGCGTTGCGCTGAGCGTGCTGTACAAATTCAGCAAGG TTGGTATGGAGAC CGTTCAGCGTGTGCTGGACATGATGGAAAAGCGTGTGGACGATGACGGCATCTTCCAAAC CTACTTTGATCCG CTGCGTCTGCGTGTTGACCCGGTGGCGGCGACCAACACCGTTTACCTGTTCCATCTGGGT GGCCGTCCGGGTC CGACCCAGCAAACCGAGCAGTTTCTGAGCAAACTGCTGGAAAAGCGTAGCTATCTGCGTG GCACCCTGTACTA TCGTACCCCGGAGGCGTTCCTGTACCAACTGACCCGTCTGGTGGTTAGCTTCGAGGAATA TTTTCGTAAAACC GGTTTCCTGGAAATGCTGAAGAAACGTCTGAGCGAGCGTGTGGGCATTGAAAGCGATGCG TTTACCCTGGCGA TGCGTATCCTGGCGTGCGTTAACTGCGGTATTCCGGTGGCGGAGCTGAACCGTGATATGG ACCGTCTGGCGCG TATGCAGAACGTTGATGGTAGCTGGGACGTGTGCCCGTACTATAGCTACAACGACCCGAA GAGCTGGTTCGGC AACGAACTGCTGACCACCGCGTTTGCGGCGGCGGCGCTGGAGCAAAGCGAACCGAACTTC AACTAA

SEQ ID NO: 3

Bazzania sp._BazzHADl_amino acid sequence

MAPPFGFIGGNATSVTTSPDVLDYNYDGGPPTGQYKSLILDIGGVLLQPNLDRSYGT IVPPVQFKRMLRSHIW FNYSMGKMSAEEVFRQLSVKSGYPAEDIRSFGRKARESLVPITQITDLLFKLSKESKVRL FCMTNCPAEDFEY LSQAYPKLFGLFERIFTSASTGMRKPNRNFFRHVLQETGIRASETIFVDDLVPNIMAAEA LDFTAIHVSPESP AETVRLLMFHLRKPEERLAAARDYLLRNRGVDKALSLTSDGVRVQVYFDHFWAEVLNDER FLPITAAPESGT MNFFPKEERKATRDITTNAVTHVDLPDDLDSTSVALSVLYKFSKVGMETVQRVLDMMEKR VDDDGIFQTYFDP LRLRVDPVAATNTVYLFHLGGRPGPTQQTEQFLSKLLEKRSYLRGTLYYRTPEAFLYQLT RLW SFEEYFRKT GFLEMLKKRLSERVGIESDAFTLAMRILACVNCGIPVAELNRDMDRLARMQNVDGSWDVC PYYSYNDPKSWFG NELLTTAFAAAALEQSEPNFN

SEQ ID NO: 4

Bazzania sp._BazzHAD2_wt_nucleic acid sequence

ATGGAAAGACCTCATTTCGACACTCTGATCATGGACCTGGGAGGTGTTCTTGTCGAC TTCTCTCTTCAAACAT CCACATCCACTTCTGTGAAATCTCTCAAGTTTGTATTACGTTCCGCCACCTGGGCAGCTT ACGAGTGCGGCCA TATGTCAGAATCGGATTGCTATGCTGCAGTGGCTAAAGATCTTGGAAGCTCTGCAAACGC ATCTCAAGTTGGT GAAGCCATCTCAGAAGTTCGAAAGTCACTGCAGGTCAATGAAGATTTGATAGGGGTGATT CGGGAGCTCAAAG CCCAGAACGGCCTTCGAGTTTACGTAATGTCCAACATACCACAACCAGACTTCGATGCCG TCAAAGCTAAGTC AGCGAGCTGGGGAGTATTCGATGGCATGTATCCCTCGTATGCGGTGGGCACCCATAAGCC CGATCTTGCTTTC TATCGACACGTACTCGAAGAGACAAACACAGACCCTCTCAAAGCGATCTTTGTCGACGAC AGCTTACAAAATG TCATTGCAGCCAGATCTTTTGGATTGACAGGAATTATCTACAATGATACTGTCAATGCTG CTCGAACGCTGCG AAATCTCCTTGGTGATCCTATCTTGAGAGGTGAGCAGTACCTTTCGTCCCACGCTGGACA ATTGCTGTCTATC TCAGACACAGGAACCCCCTTCCCAGATAACTTTTCACAGCTTTTAATTTTGGATGCTACC GGAAACCGTGATC TTGTTGTTCTTGAAAACCCTCAGCGAACATGGAACTACTTCATCGGAAAGCCTGTGTTGA CGTCTGAAACATT CCCTGATGATCTAGATACGACATCTATCGCCCTCATGGCATTGAATGTAGACCTAGAGGT GGCCAATTCTGTG ATGGATGAGATGCTGGAGTTTAGAAACAGTGATGGATTGTTTCTGACGTACTTTGACGAA ACAAGGCCGCGGG TAGATGCTGTTGTCAATGTTAATGTTCTTCGTCTCTTCCATCGACATGAACGGGAGAGAG AGGCCCAGCAACC GCTGGAGTGGATTACCAACGTCCTTACCCACCGAGCGTATGTTGATGGCACACTATTTTA CTACCATGCTGAG AGCTTCTTATATTTCCTTTCGCGACTGTTCTGTGAGAACTCCAGCGTCCAATCACGATTT CAAGAGCTTCTTG AACAACGCCTGCGGGAACGAATTGGAACCCCGGGCGACGCACTGAGTCTAGCCATGCGTG CATTAGCATGCCA GATGCTGGGAATCGATAGTTCTACCGACGTTCGCGCTCTTCTGCCCTTGCAATGCGACGA CGGCGGTTGGGAA GCTGGCTGGGTGTGCAGATACGGTTCAAATGGTATGCGCGTTGGAAGCCGGGGATACACT ACGGCTTTGGCTA TAAATGCTATCAGAGGGGCAAGAGTCTCGAGGTGA

SEQ ID NO: 5

Bazzania sp._BazzHAD2_E. coll optimized

ATGGAACGTCCGCACTTCGACACCCTGATCATGGATCTGGGTGGCGTGCTGGTTGAC TTTAGCCTGCAGACCA GCACCAGCACCAGCGTTAAGAGCCTGAAATTTGTGCTGCGTAGCGCGACCTGGGCGGCGT ACGAATGCGGTCA CATGAGCGAGAGCGACTGCTATGCGGCGGTTGCGAAGGATCTGGGTAGCAGCGCGAACGC GAGCCAAGTGGGT GAAGCGATCAGCGAGGTTCGTAAGAGCCTGCAAGTGAACGAAGATCTGATCGGTGTTATT CGTGAGCTGAAAG CGCAGAACGGCCTGCGTGTGTACGTTATGAGCAACATTCCGCAACCGGACTTCGATGCGG TTAAGGCGAAAAG CGCGAGCTGGGGTGTGTTTGATGGCATGTACCCGAGCTATGCGGTTGGCACCCACAAGCC GGACCTGGCGTTT TATCGTCACGTGCTGGAGGAAACCAACACCGATCCGCTGAAAGCGATCTTCGTTGACGAT AGCCTGCAAAACG TGATTGCGGCGCGTAGCTTTGGTCTGACCGGCATCATTTACAACGACACCGTGAACGCGG CGCGTACCCTGCG TAACCTGCTGGGTGATCCGATCCTGCGTGGCGAACAGTATCTGAGCAGCCACGCGGGTCA ACTGCTGAGCATT AGCGACACCGGCACCCCGTTCCCGGATAACTTTAGCCAGCTGCTGATTCTGGACGCGACC GGTAACCGTGATC TGGTGGTTCTGGAAAACCCGCAACGTACCTGGAACTACTTCATCGGCAAACCGGTTCTGA CCAGCGAGACCTT TCCGGACGATCTGGACACCACCAGCATTGCGCTGATGGCGCTGAACGTGGACCTGGAAGT TGCGAACAGCGTG ATGGATGAAATGCTGGAGTTCCGTAACAGCGACGGTCTGTTCCTGACCTACTTTGACGAG ACCCGTCCGCGTG TGGATGCGGTGGTTAACGTGAACGTTCTGCGTCTGTTTCACCGTCACGAGCGTGAACGTG AGGCGCAGCAACC GCTGGAGTGGATTACCAACGTTCTGACCCACCGTGCGTACGTGGACGGCACCCTGTTCTA CTATCACGCGGAA AGCTTCCTGTATTTTCTGAGCCGTCTGTTCTGCGAGAACAGCAGCGTTCAGAGCCGTTTT CAAGAACTGCTGG AGCAGCGTCTGCGTGAACGTATTGGCACCCCGGGTGATGCGCTGAGCCTGGCGATGCGTG CGCTGGCGTGCCA GATGCTGGGTATTGACAGCAGCACCGATGTGCGTGCGCTGCTGCCGCTGCAATGCGATGA TGGTGGCTGGGAG GCGGGTTGGGTTTGCCGTTACGGTAGCAACGGCATGCGTGTGGGTAGCCGTGGCTATACC ACCGCGCTGGCGA TCAACGCGATTCGTGGTGCGCGTGTTAGCCGTTAA

SEQ ID NO: 6

Bazzania sp ._BazzHAD2_amino acid sequence

MERPHFDTLIMDLGGVLVDFSLQTSTSTSVKSLKFVLRSATWAAYECGHMSESDCYA AVAKDLGSSANASQVG EAISEVRKSLQVNEDLIGVIRELKAQNGLRVYVMSNIPQPDFDAVKAKSASWGVFDGMYP SYAVGTHKPDLAF YRHVLEETNTDPLKAIFVDDSLQNVIAARSFGLTGIIYNDTVNAARTLRNLLGDPILRGE QYLSSHAGQLLSI SDTGTPFPDNFSQLLILDATGNRDLW LENPQRTWNYFIGKPVLTSETFPDDLDTTSIALMALNVDLEVANSV MDEMLEFRNSDGLFLTYFDETRPRVDAWNVNVLRLFHRHEREREAQQPLEWITNVLTHRA YVDGTLFYYHAE SFLYFLSRLFCENSSVQSRFQELLEQRLRERIGTPGDALSLAMRALACQMLGIDSSTDVR ALLPLQCDDGGWE AGWVCRYGSNGMRVGSRGYTTALAINAIRGARVSR SEQ ID NO: 7

Bazzania sp _BazzHAD3_wt_nucleic acid sequence

ATGCCTGCTTTAAGCTCGCATATGGAGCCACCTAATTTTGACACCCTGATCCTGGAC CTGGGAGATGTCCTTG TGGGAGGCTCTCTTCAAGGCTCTTACGCCTTGGCCATGAAGAGAATTCTGAAACTCTCAT TACGTACCCCGAC GTGGGGAAAATACGAGTGCGGCCAATTATCAGAATTAGATTGCTATGACGCTGTGGCTGA AGATCTTGGGAAC TCCACAAGCTCCGCTCAAGTGGGTGAGGTCGTCTCAGAAGCTCGAAAGTCATTGCAGGTC AATGAAGACCTGA TAAAGGTGCTTCAGGAGCTAAAGGCTGAGAACAATCTCCGGGTTTATGTCATGTCCAATA TACCCAAGCCGGA TTTGGCTGTTGTGAAGGCCAAGTCAGTGAACTGGGGAGTCATCGATGGCTGGTATCCCTC TTATGCTGTGGGC TTCCACAAGCCGGATCTCGCTTTCTACCGGCATGTACTTGAAGAGACAAACACCGATCCT CTTAAGGCCATCT TCGTTGATGACAAGGTTCAGAATGTCATTGTTGCCAGATCCTTTGGAATGACAGGCATTG TCTTCAAGGACAC TAAGAGCACTACCAAGGAGCTGAAGAATCTCCTTGGTGACCCCGTCAAGAGAGGGGAGCA CTTCCTCTCAGCT CATGCCAAACAATTGGAGTCTGTCTGTGGGGATGGCACCACTTTCCTTGACAACTTCGCC CAGCTGCTGATTC TTCATGATACTGGAAACAGCGATCTTGTGGCTCGTCAGCACCATGGCAGAACCTGGAACT ACTTCATTGGCAA GCCGATGGGGACGACAGACACATTCCCCAACGACCTAGATACCACCTCCATCGCTCTGCT GACTCTGAATGTA GACTCAGAAGTAGCGAGGTCTGTGTTAGATGAGATGCTGCTGTACACCAGCAGTGATGGG CTGGTAGAGGTGT ATTTTGACAAAACTAGACCTCGGGTGGATCCCGTTGTCTGTGTCAACGTGCTACGTCTCT TCTCTAAATATGG GAGGGAGCTCCAGCTTCAGAAGACTCTGGACTGGGTAACTGAGGTCCTTGTTCACCGAGC TTACATTGACGGC ACACTCTTCTACTACCACGCTGAGAGCTTCTTGTACTTCCTTTCGTGTCTCTACAAAGAG AACCCCCGGCTGC AGACTCAATTTCGGGAGCCTCTACAAGAACGCCTGCGGGAGAGGATTGGGCAGCCCGGGG ATGCACTTTGTCT GGCTATGCGTGCAATCGCATGCCAGACTGTGGGGATTGGGAATGTTATTGATGTGCAAGC TCTTCTGCCATTG CAATCTAGTGATGGTGGTTGGGAGGCTGGCTGGGTCTGCAGAATGGGCACGTCTGGTGTT CCTGTTGGAAACC GAGGAGTCACAACGGCACTTTGTATTAGAGCTATTCGAGGCGCATCAGGGAGCTACCACA CTTGA

SEQ ID NO: 8

Bazzania sp._BazzHAD3_E. coll optimized

ATGCCGGCGCTGAGCAGCCACATGGAACCGCCGAACTTCGACACCCTGATTCTGGAC CTGGGTGATGTGCTGG TTGGTGGCAGCCTGCAGGGTAGCTACGCGCTGGCGATGAAGCGTATCCTGAAACTGAGCC TGCGTACCCCGAC CTGGGGCAAGTACGAATGCGGCCAGCTGAGCGAGCTGGACTGCTATGATGCGGTTGCGGA AGACCTGGGTAAC AGCACCAGCAGCGCGCAAGTGGGCGAGGTGGTTAGCGAAGCGCGTAAAAGCCTGCAGGTT AACGAGGATCTGA TCAAGGTGCTGCAAGAGCTGAAAGCGGAAAACAACCTGCGTGTGTACGTTATGAGCAACA TTCCGAAGCCGGA CCTGGCGGTGGTTAAGGCGAAAAGCGTGAACTGGGGTGTTATCGATGGCTGGTATCCGAG CTATGCGGTTGGT TTCCACAAACCGGACCTGGCGTTTTATCGTCACGTGCTGGAGGAAACCAACACCGATCCG CTGAAGGCGATCT TCGTGGACGATAAAGTTCAGAACGTGATTGTTGCGCGTAGCTTCGGTATGACCGGCATCG TTTTTAAGGACAC CAAAAGCACCACCAAGGAACTGAAAAACCTGCTGGGTGATCCGGTTAAGCGTGGCGAACA CTTTCTGAGCGCG CACGCGAAACAACTGGAGAGCGTGTGCGGTGACGGCACCACCTTCCTGGATAACTTTGCG CAGCTGCTGATTC TGCACGACACCGGTAACAGCGATCTGGTGGCGCGTCAACACCACGGCCGTACCTGGAACT ACTTCATTGGCAA GCCGATGGGCACCACCGATACCTTTCCGAACGACCTGGATACCACCAGCATCGCGCTGCT GACCCTGAACGTG GACAGCGAAGTTGCGCGTAGCGTGCTGGATGAGATGCTGCTGTACACCAGCAGCGACGGT CTGGTTGAGGTGT ATTTCGACAAAACCCGTCCGCGTGTTGATCCGGTGGTTTGCGTGAACGTTCTGCGTCTGT TTAGCAAGTACGG TCGTGAACTGCAGCTGCAAAAAACCCTGGATTGGGTGACCGAGGTGCTGGTTCACCGTGC GTATATCGACGGC ACCCTGTTCTACTATCACGCGGAAAGCTTCCTGTACTTTCTGAGCTGCCTGTATAAGGAG AACCCGCGTCTGC AGACCCAATTTCGTGAGCCGCTGCAGGAACGTCTGCGTGAGCGTATTGGTCAGCCGGGTG ATGCGCTGTGCCT GGCGATGCGTGCGATCGCGTGCCAGACCGTTGGTATCGGCAACGTGATTGATGTTCAAGC GCTGCTGCCGCTG CAGAGCAGCGATGGTGGCTGGGAGGCGGGTTGGGTTTGCCGTATGGGCACCAGCGGTGTG CCGGTTGGTAACC GTGGCGTGACCACCGCGCTGTGCATCCGTGCGATTCGTGGTGCGAGCGGCAGCTATCACA CCTAA

SEQ ID NO: 9

Bazzania sp._BazzHAD3_amino acid sequence

MPALSSHMEPPNFDTLILDLGDVLVGGSLQGSYALAMKRILKLSLRTPTWGKYECGQ LSELDCYDAVAEDLGN STSSAQVGEW SEARKSLQVNEDLIKVLQELKAENNLRVYVMSNIPKPDLAW KAKSVNWGVIDGWYPSYAVG FHKPDLAFYRHVLEETNTDPLKAIFVDDKVQNVIVARSFGMTGIVFKDTKSTTKELKNLL GDPVKRGEHFLSA HAKQLESVCGDGTTFLDNFAQLLILHDTGNSDLVARQHHGRTWNYFIGKPMGTTDTFPND LDTTSIALLTLNV DSEVARSVLDEMLLYTSSDGLVEVYFDKTRPRVDPW CVNVLRLFSKYGRELQLQKTLDWVTEVLVHRAYIDG TLFYYHAESFLYFLSCLYKENPRLQTQFREPLQERLRERIGQPGDALCLAMRAIACQTVG IGNVIDVQALLPL QSSDGGWEAGWVCRMGTSGVPVGNRGVTTALCIRAIRGASGSYHT

SEQ ID NO: 10 Bazzania trilobata _BtHAD_wt_nucleic acid sequence

ATGGCTCCTCCTTTCGGATATATTGGTGGGAATGAATCGAGTGTGACGACGTCCCCA GACGTCCTCGACTACA GCTATGATGGTGGCCCTCCTACTGGCCAATACAAGAGTATGATTTTAGACATTGGCGGAG TGTTGCTTCAGCC CAACCTTGACAGGAGCTATGGCACGATTGTACCACCTGTGCAATTCAAACGTATGTTGCG CTCGCACATCTGG TTCAATTACTCAATGGGAAAGGTGTCTGCGGAGGAAGTGTTTCGGCAGCTCTCCGTGAAG TCTGGCTACCCGG CAGAAGATATCCGGAGTTTTGCTCGAAAAGCGAGAGAGTCTCTTGTGCCCATCACACAAA TAACGGATCTTCT GGTCAAACTCAGCAAGGAATCGAAGATCAGACTGTTCTGTATGACCAATTGCCCTGCCGA AGACTTCGCATAC CTCTCACAGGCATATCCCAACCTTTTCGGATTGTTCGAGAGGATTTTTACGTCGGCGTCC ACCGGTATGCGCA AGCCAAACAGGAATTTCTTCCGACATGTACTCCAGGAGACGGGAATCAGTGCGACGGAGA CTATTTTCGTAGA CGACCTGGTACCGAACATCATGGCCGCCGAGGCGCTGGACTTCACGGCAATTCACGTTTC TCCAGAGTCGCCC GCCGAGACCGTGAGGTTGCTGATGTTCCATCTCCGTAAGCCAGAAGAGCGGCTGGCGGCG GCGCGCGACTATC TCCTGCGCAACCGTGGAGTGGACAAAGCTCTCTCTCTCACATCTGACGGAGTCCGAGTCC AGGTTTATTTCGA CCACTTCGTGGTCGCCGAAGTGCTCAACGACGAGCGATTCTTGCCAATCACTGCAGCTCC GAAGTCCGGGACT ATGAACTTTTTCCCGAAGGAGGAACGCAACGCGACTCGAGATATGACGACCAATGCAGTC ACACACGTGGACC TTCCCGATGATCTTGACTCCACCTCTGTCGCCTTATCGGTCCTTTACAAATTCTCCAAAG TGGCGATGGAGAC GGTTCAGAGGGTCCTGGATATGATGGAGAAACGTGTGGACGATGACGGCATTTTTCAAAC GTACTTCGATCCT TTGCGGCTGCGTGTTGACCCGGTGGCCGCTACTAACACCGTCTACCTCTTCCATTTAGGA GGTCGTCCCGGAC CTACTCAGCAGACGGAACAATTCCTCTCCAAGTTACTGGAAAAGAGATCTTACGTCCGCG GCACACTCTACTA CAGGACCCCCGAGGCATTTCTCTATCAACTCACCAGATTGGTGGTATCATTTGAGTATTT CAGAAAGACAGGC TTCCTGGAAATGCTCAAGAAAAGGCTGAGTGAGCGAGTGGGCATCGAGAGCGATGCATTC ACGTTGGCGATGA GGATTCTAGCGTGTGTGAATTGTGGAATCCCTGTTGCTGAGCTGAACAGAGATATGGACA GACTAGCACGGAT GCAAAATGTGGATGGTTCATGGGACGTCTGCCCTTACTACAGCTACAATGATCCCAAGAG TTGGTTTGGCAAC GAGCTTCTTACCACAGCGTTTGCTGCTGCCGCTCTTGAGCATTCTGAGCCCAACTTTAAG TGA

SEQ ID NO: 11

Bazzania trilobata _BtHAD_E. coll optimized

ATGGCTCCGCCGTTCGGTTACATTGGTGGCAACGAGAGCAGCGTTACCACCAGCCCG GATGTGCTGGACTACA GCTATGATGGTGGCCCGCCGACCGGCCAGTACAAAAGCATGATCCTGGATATTGGTGGCG TTCTGCTGCAACC GAACCTGGACCGTAGCTATGGCACCATCGTTCCGCCGGTGCAGTTCAAGCGTATGCTGCG TAGCCACATTTGG TTTAACTACAGCATGGGTAAAGTTAGCGCGGAGGAAGTGTTCCGTCAACTGAGCGTGAAG AGCGGCTATCCGG CGGAGGATATCCGTAGCTTTGCGCGTAAAGCGCGTGAAAGCCTGGTTCCGATCACCCAGA TTACCGACCTGCT GGTGAAGCTGAGCAAAGAGAGCAAGATTCGTCTGTTCTGCATGACCAACTGCCCGGCGGA AGACTTTGCGTAC CTGAGCCAAGCGTATCCGAACCTGTTCGGTCTGTTTGAGCGTATCTTCACCAGCGCGAGC ACCGGCATGCGTA AGCCGAACCGTAACTTCTTTCGTCACGTTCTGCAGGAGACCGGCATCAGCGCGACCGAAA CCATTTTCGTTGA CGATCTGGTGCCGAACATCATGGCGGCGGAAGCGCTGGATTTTACCGCGATTCACGTTAG CCCGGAGAGCCCG GCGGAAACCGTGCGTCTGCTGATGTTTCACCTGCGTAAACCGGAGGAACGTCTGGCGGCG GCGCGTGACTACC TGCTGCGTAACCGTGGTGTTGATAAAGCGCTGAGCCTGACCAGCGATGGTGTGCGTGTTC AAGTGTATTTCGA TCACTTTGTGGTTGCGGAAGTGCTGAACGACGAACGTTTCCTGCCGATTACCGCGGCGCC GAAAAGCGGCACC ATGAACTTCTTTCCGAAAGAGGAGCGTAACGCGACCCGTGATATGACCACCAACGCGGTT ACCCACGTGGATC TGCCGGACGATCTGGACAGCACCAGCGTTGCGCTGAGCGTGCTGTACAAATTTAGCAAGG TTGCGATGGAGAC CGTTCAGCGTGTGCTGGACATGATGGAAAAACGTGTGGACGATGACGGCATCTTCCAAAC CTACTTTGATCCG CTGCGTCTGCGTGTTGACCCGGTGGCGGCGACCAACACCGTTTACCTGTTCCATCTGGGT GGCCGTCCGGGTC CGACCCAGCAAACCGAGCAGTTTCTGAGCAAACTGCTGGAAAAGCGTAGCTATGTGCGTG GCACCCTGTACTA TCGTACCCCGGAAGCGTTCCTGTACCAACTGACCCGTCTGGTGGTTAGCTTCGAGTATTT TCGTAAGACCGGT TTTCTGGAAATGCTGAAGAAACGTCTGAGCGAGCGTGTGGGCATCGAAAGCGATGCGTTC ACCCTGGCGATGC GTATCCTGGCGTGCGTTAACTGCGGTATTCCGGTGGCGGAGCTGAACCGTGATATGGACC GTCTGGCGCGTAT GCAGAACGTTGATGGTAGCTGGGACGTGTGCCCGTACTATAGCTACAACGACCCGAAAAG CTGGTTCGGCAAC GAACTGCTGACCACCGCGTTTGCGGCGGCGGCGCTGGAGCACAGCGAACCGAACTTCAAG TAA

SEQ ID NO: 12

Bazzania trilobata _BtHAD_amino acid sequence

MAPPFGYIGGNESSVTTSPDVLDYSYDGGPPTGQYKSMILDIGGVLLQPNLDRSYGT IVPPVQFKRMLRSHIW FNYSMGKVSAEEVFRQLSVKSGYPAEDIRSFARKARESLVPITQITDLLVKLSKESKIRL FCMTNCPAEDFAY LSQAYPNLFGLFERIFTSASTGMRKPNRNFFRHVLQETGISATETIFVDDLVPNIMAAEA LDFTAIHVSPESP AETVRLLMFHLRKPEERLAAARDYLLRNRGVDKALSLTSDGVRVQVYFDHFWAEVLNDER FLPITAAPKSGT MNFFPKEERNATRDMTTNAVTHVDLPDDLDSTSVALSVLYKFSKVAMETVQRVLDMMEKR VDDDGIFQTYFDP LRLRVDPVAATNTVYLFHLGGRPGPTQQTEQFLSKLLEKRSYVRGTLYYRTPEAFLYQLT RLW SFEYFRKTG FLEMLKKRLSERVGIESDAFTLAMRILACVNCGIPVAELNRDMDRLARMQNVDGSWDVCP YYSYNDPKSWFGN

ELLTTAFAAAALEHSEPNFK

SEQ ID NO: 13

Artificial _BtHAD mutant of class II motif_E.coli optimized

ATGGCTCCGCCGTTCGGTTACATTGGTGGCAACGAGAGCAGCGTTACCACCAGCCCG GATGTGCTGGACTACA GCTATGATGGTGGCCCGCCGACCGGCCAGTACAAAAGCATGATCCTGGATATTGGTGGCG TTCTGCTGCAACC GAACCTGGACCGTAGCTATGGCACCATCGTTCCGCCGGTGCAGTTCAAGCGTATGCTGCG TAGCCACATTTGG TTTAACTACAGCATGGGTAAAGTTAGCGCGGAGGAAGTGTTCCGTCAACTGAGCGTGAAG AGCGGCTATCCGG CGGAGGATATCCGTAGCTTTGCGCGTAAAGCGCGTGAAAGCCTGGTTCCGATCACCCAGA TTACCGACCTGCT GGTGAAGCTGAGCAAAGAGAGCAAGATTCGTCTGTTCTGCATGACCAACTGCCCGGCGGA AGACTTTGCGTAC CTGAGCCAAGCGTATCCGAACCTGTTCGGTCTGTTTGAGCGTATCTTCACCAGCGCGAGC ACCGGCATGCGTA AGCCGAACCGTAACTTCTTTCGTCACGTTCTGCAGGAGACCGGCATCAGCGCGACCGAAA CCATTTTCGTTGA CGATCTGGTGCCGAACATCATGGCGGCGGAAGCGCTGGATTTTACCGCGATTCACGTTAG CCCGGAGAGCCCG GCGGAAACCGTGCGTCTGCTGATGTTTCACCTGCGTAAACCGGAGGAACGTCTGGCGGCG GCGCGTGACTACC TGCTGCGTAACCGTGGTGTTGATAAAGCGCTGAGCCTGACCAGCGATGGTGTGCGTGTTC AAGTGTATTTCGA TCACTTTGTGGTTGCGGAAGTGCTGAACGACGAACGTTTCCTGCCGATTACCGCGGCGCC GAAAAGCGGCACC ATGAACTTCTTTCCGAAAGAGGAGCGTAACGCGACCCGTGATATGACCACCAACGCGGTT ACCCACGTGGATC TGCCGGACGCGCTGGCGAGCACCAGCGTTGCGCTGAGCGTGCTGTACAAATTTAGCAAGG TTGCGATGGAGAC CGTTCAGCGTGTGCTGGACATGATGGAAAAACGTGTGGACGATGACGGCATCTTCCAAAC CTACTTTGATCCG CTGCGTCTGCGTGTTGACCCGGTGGCGGCGACCAACACCGTTTACCTGTTCCATCTGGGT GGCCGTCCGGGTC CGACCCAGCAAACCGAGCAGTTTCTGAGCAAACTGCTGGAAAAGCGTAGCTATGTGCGTG GCACCCTGTACTA TCGTACCCCGGAAGCGTTCCTGTACCAACTGACCCGTCTGGTGGTTAGCTTCGAGTATTT TCGTAAGACCGGT TTTCTGGAAATGCTGAAGAAACGTCTGAGCGAGCGTGTGGGCATCGAAAGCGATGCGTTC ACCCTGGCGATGC GTATCCTGGCGTGCGTTAACTGCGGTATTCCGGTGGCGGAGCTGAACCGTGATATGGACC GTCTGGCGCGTAT GCAGAACGTTGATGGTAGCTGGGACGTGTGCCCGTACTATAGCTACAACGACCCGAAAAG CTGGTTCGGCAAC GAACTGCTGACCACCGCGTTTGCGGCGGCGGCGCTGGAGCACAGCGAACCGAACTTCAAG TAA

SEQ ID NO: 14

Artificial _BtHAD mutant of class II motif

MAPPFGYIGGNESSVTTSPDVLDYSYDGGPPTGQYKSMILDIGGVLLQPNLDRSYGT IVPPVQFKRMLRSHIW FNYSMGKVSAEEVFRQLSVKSGYPAEDIRSFARKARESLVPITQITDLLVKLSKESKIRL FCMTNCPAEDFAY LSQAYPNLFGLFERIFTSASTGMRKPNRNFFRHVLQETGISATETIFVDDLVPNIMAAEA LDFTAIHVSPESP AETVRLLMFHLRKPEERLAAARDYLLRNRGVDKALSLTSDGVRVQVYFDHFWAEVLNDER FLPITAAPKSGT MNFFPKEERNATRDMTTNAVTHVDLPDALASTSVALSVLYKFSKVAMETVQRVLDMMEKR VDDDGIFQTYFDP LRLRVDPVAATNTVYLFHLGGRPGPTQQTEQFLSKLLEKRSYVRGTLYYRTPEAFLYQLT RLW SFEYFRKTG FLEMLKKRLSERVGIESDAFTLAMRILACVNCGIPVAELNRDMDRLARMQNVDGSWDVCP YYSYNDPKSWFGN ELLTTAFAAAALEHSEPNFK

SEQ ID NO: 15

Artificial _BtHAD mutant of QW motif_F. coli optimized

ATGGCTCCGCCGTTCGGTTACATTGGTGGCAACGAGAGCAGCGTTACCACCAGCCCG GATGTGCTGGACTACA GCTATGATGGTGGCCCGCCGACCGGCCAGTACAAAAGCATGATCCTGGATATTGGTGGCG TTCTGCTGCAACC GAACCTGGACCGTAGCTATGGCACCATCGTTCCGCCGGTGCAGTTCAAGCGTATGCTGCG TAGCCACATTTGG TTTAACTACAGCATGGGTAAAGTTAGCGCGGAGGAAGTGTTCCGTCAACTGAGCGTGAAG AGCGGCTATCCGG CGGAGGATATCCGTAGCTTTGCGCGTAAAGCGCGTGAAAGCCTGGTTCCGATCACCCAGA TTACCGACCTGCT GGTGAAGCTGAGCAAAGAGAGCAAGATTCGTCTGTTCTGCATGACCAACTGCCCGGCGGA AGACTTTGCGTAC CTGAGCCAAGCGTATCCGAACCTGTTCGGTCTGTTTGAGCGTATCTTCACCAGCGCGAGC ACCGGCATGCGTA AGCCGAACCGTAACTTCTTTCGTCACGTTCTGCAGGAGACCGGCATCAGCGCGACCGAAA CCATTTTCGTTGA CGATCTGGTGCCGAACATCATGGCGGCGGAAGCGCTGGATTTTACCGCGATTCACGTTAG CCCGGAGAGCCCG GCGGAAACCGTGCGTCTGCTGATGTTTCACCTGCGTAAACCGGAGGAACGTCTGGCGGCG GCGCGTGACTACC TGCTGCGTAACCGTGGTGTTGATAAAGCGCTGAGCCTGACCAGCGATGGTGTGCGTGTTC AAGTGTATTTCGA TCACTTTGTGGTTGCGGAAGTGCTGAACGACGAACGTTTCCTGCCGATTACCGCGGCGCC GAAAAGCGGCACC ATGAACTTCTTTCCGAAAGAGGAGCGTAACGCGACCCGTGATATGACCACCAACGCGGTT ACCCACGTGGATC TGCCGGACGATCTGGACAGCACCAGCGTTGCGCTGAGCGTGCTGTACAAATTTAGCAAGG TTGCGATGGAGAC CGTTCAGCGTGTGCTGGACATGATGGAAAAACGTGTGGACGATGACGGCATCTTCCAAAC CTACTTTGATCCG CTGCGTCTGCGTGTTGACCCGGTGGCGGCGACCAACACCGTTTACCTGTTCCATCTGGGT GGCCGTCCGGGTC CGACCCAGCAAACCGAGCAGTTTCTGAGCAAACTGCTGGAAAAGCGTAGCTATGTGCGTG GCACCCTGTACTA TCGTACCCCGGAAGCGTTCCTGTACCAACTGACCCGTCTGGTGGTTAGCTTCGAGTATTT TCGTAAGACCGGT TTTCTGGAAATGCTGAAGAAACGTCTGAGCGAGCGTGTGGGCATCGAAAGCGATGCGTTC ACCCTGGCGATGC GTATCCTGGCGTGCGTTAACTGCGGTATTCCGGTGGCGGAGCTGAACCGTGATATGGACC GTCTGGCGCGTAT GCAGAACGTTGCGGGTAGCTGGGACGTGTGCCCGTACTATAGCTACAACGACCCGAAAAG CTGGTTCGGCAAC GAACTGCTGACCACCGCGTTTGCGGCGGCGGCGCTGGAGCACAGCGAACCGAACTTCAAG TAA

SEQ ID NO: 16

Artificial_BtHAD mutant of QW motif

MAPPFGYIGGNESSVTTSPDVLDYSYDGGPPTGQYKSMILDIGGVLLQPNLDRSYGT IVPPVQFKRMLRSHIW FNYSMGKVSAEEVFRQLSVKSGYPAEDIRSFARKARESLVPITQITDLLVKLSKESKIRL FCMTNCPAEDFAY LSQAYPNLFGLFERIFTSASTGMRKPNRNFFRHVLQETGISATETIFVDDLVPNIMAAEA LDFTAIHVSPESP AETVRLLMFHLRKPEERLAAARDYLLRNRGVDKALSLTSDGVRVQVYFDHFWAEVLNDER FLPITAAPKSGT MNFFPKEERNATRDMTTNAVTHVDLPDDLDSTSVALSVLYKFSKVAMETVQRVLDMMEKR VDDDGIFQTYFDP LRLRVDPVAATNTVYLFHLGGRPGPTQQTEQFLSKLLEKRSYVRGTLYYRTPEAFLYQLT RLW SFEYFRKTG FLEMLKKRLSERVGIESDAFTLAMRILACVNCGIPVAELNRDMDRLARMQNVAGSWDVCP YYSYNDPKSWFGN ELLTTAFAAAALEHSEPNFK

SEQ ID NO: 17

Selaginella moellendorffii_SmHADl_wt_nucleic acid sequence

ATGATAATCATTAGCTTTGCTTGCTTGCTCAAGTTTGGATGTGGAGACAATGGCCCC CGGGGAGATCTCCTCC GCAGGGCTCTCCAGCACTCCTCCTTCTTGGCCTATTCTTGCGGGGAGCTCGATCGCACTG CCGCAATCTCGAC AATCTCGCGCAAATTCAAGCTCACGGACCCAGCTCTCGTGGATTCCATGCTGCTCGAGGC CGCTTCCAGCTGC GAGGTGGACAAGGAGCTACTGTCACTGCTCCAGTCAACGAGGCAGCAGCTCCTGGGATGG ATTGATATTCCTC CACAAGAGTGGGAGCGAGTTTACAGCCTCTTCCCGGTGTCGCTCTGGAAGAACTTCGCTA CAATCTCCCGCGA TCTAGATTCCCTCCTTGGAGATATCAGGTCCCATGCTGTGATTGTCGATAAAAGCGTGGA GATGGCCGCGCTA CATGCCTTCGAGAGCTTGCTCGCTCTCCCGTATGCCTCGCCCAACAAGAAAGCTTGCCAG GACTTCTTGAAGC GTCGGTTCCTGCTGCCTCGAGCTGTGAAGCTGGGGATGGATCGAGTCAAGGAGGAGGTGA GATCCAAGCAGCT CAAATCACTCTACCTGCTTGACAATGGCCGGCACGAGCTCGTCTCCGAGATCTTCTTTCC CTGCGTGGCGGCC TGGTGCCTTCCGGAGATCATTCCTCTCGGCTGGATGAAATATCTCCAGGTACTGATCAAG CGCGGCCATCCCT TCGGTTACTTCGGTGCCGACGTCTCGCGCTATCCGCCGGACATCGACACCATGTCGACTT GTGTCTCCACACT TTTTGATCTTAGTCTCGTCACCTCCGCGCAAGCTATGCACTTTCTGGAGATTTGTCTCGA GAACGTCAACGAT CAAAACCAGCTTCTCACGTACCTGGACGTGGAGCGGCCTCGAGTGGATCCGGTCGTCATA GCCAACGTTGTCT ATCTCGCCTATGCGCTCTCCATGGAAGATCACCCGGTTGTCCGCCACAACGAGAATCTGA TCCAGAGATATCT GTTAAGCGGCGGATTCGTATACGGGACTCGCTATTACCTCAGCCAGGAGGACTTCTTGTT CATGTATGGCCGG GTCCTCGCGACGTTTGGAGAGAAGAGGGAGATTCCCAACTTTGATCTCGTGTATCAAGCG ATGGAGGCGGCTC TCGTCAACCGGATCGGGAACGAGACCGAGTCGAAGCCACTGGACGTTGCCAAAAGGATTC TGCTCAGCAGGGG TTTTGGAATCCGGAACACGATAGATGTAGACTTGCTCCTCAAGATGCAGAACGAGGATGG TTCCTGGCCTTTG CAAGTGCTCAGCAATCTTCCATCTGCTAAAGGTGGTGTCTTCAATTCGGTGGTGGACTTG AGCTTTGCGGTGA GAGCACTGCAAAGTCAAGATTGA

SEQ ID NO: 18

Selaginella moellendorffli_SmHAOl_E. coll optimized

ATGATCATTATCAGCTTCGCGTGCCTGCTGAAGTTTGGTTGCGGTGACAACGGTCCG CGTGGCGATCTGCTGC GTCGTGCGCTGCAGCACAGCAGCTTCCTGGCGTATAGCTGCGGCGAGCTGGACCGTACCG CGGCGATTAGCAC CATCAGCCGTAAGTTTAAACTGACCGACCCGGCGCTGGTTGATAGCATGCTGCTGGAAGC GGCGAGCAGCTGC GAGGTGGACAAAGAACTGCTGAGCCTGCTGCAGAGCACCCGTCAGCAACTGCTGGGTTGG ATTGATATCCCGC CGCAAGAGTGGGAACGTGTGTACAGCCTGTTCCCGGTTAGCCTGTGGAAGAACTTTGCGA CCATTAGCCGTGA CCTGGATAGCCTGCTGGGCGACATTCGTAGCCACGCGGTGATCGTTGATAAAAGCGTTGA GATGGCGGCGCTG CACGCGTTCGAAAGCCTGCTGGCGCTGCCGTATGCGAGCCCGAACAAGAAAGCGTGCCAG GACTTCCTGAAGC GTCGTTTTCTGCTGCCGCGTGCGGTGAAGCTGGGTATGGATCGTGTGAAAGAGGAAGTTC GTAGCAAGCAACT GAAAAGCCTGTACCTGCTGGACAACGGCCGTCACGAGCTGGTTAGCGAAATTTTCTTTCC GTGCGTTGCGGCG TGGTGCCTGCCGGAGATTATCCCGCTGGGTTGGATGAAGTATCTGCAGGTTCTGATTAAA CGTGGCCACCCGT TCGGTTACTTTGGTGCGGATGTGAGCCGTTATCCGCCGGACATCGATACCATGAGCACCT GCGTGAGCACCCT GTTCGATCTGAGCCTGGTTACCAGCGCGCAAGCGATGCACTTTCTGGAGATCTGCCTGGA AAACGTTAACGAC CAGAACCAACTGCTGACCTACCTGGACGTGGAACGTCCGCGTGTTGATCCGGTGGTTATT GCGAACGTGGTTT ACCTGGCGTATGCGCTGAGCATGGAGGATCACCCGGTGGTTCGTCACAACGAAAACCTGA TCCAGCGTTACCT GCTGAGCGGTGGCTTCGTTTATGGCACCCGTTACTATCTGAGCCAAGAGGACTTCCTGTT TATGTACGGTCGT GTGCTGGCGACCTTCGGCGAGAAACGTGAAATCCCGAACTTTGATCTGGTGTATCAGGCG ATGGAAGCGGCGC TGGTTAACCGTATTGGTAACGAGACCGAAAGCAAGCCGCTGGACGTTGCGAAACGTATCC TGCTGAGCCGTGG TTTTGGCATTCGTAACACCATCGACGTGGATCTGCTGCTGAAGATGCAGAACGAGGATGG CAGCTGGCCGCTG CAAGTGCTGAGCAACCTGCCGAGCGCGAAAGGTGGCGTTTTCAACAGCGTGGTTGACCTG AGCTTTGCGGTGC GTGCGCTGCAGAGCCAAGATTAA

SEQ ID NO: 19

Selaginella moellendorffii_SmHAOl_a ino acid sequence

MIIISFACLLKFGCGDNGPRGDLLRRALQHSSFLAYSCGELDRTAAISTISRKFKLT DPALVDSMLLEAASSC EVDKELLSLLQSTRQQLLGWIDIPPQEWERVYSLFPVSLWKNFATISRDLDSLLGDIRSH AVIVDKSVEMAAL HAFESLLALPYASPNKKACQDFLKRRFLLPRAVKLGMDRVKEEVRSKQLKSLYLLDNGRH ELVSEIFFPCVAA WCLPEIIPLGWMKYLQVLIKRGHPFGYFGADVSRYPPDIDTMSTCVSTLFDLSLVTSAQA MHFLEICLENVND QNQLLTYLDVERPRVDPW IANW YLAYALSMEDHPW RHNENLIQRYLLSGGFVYGTRYYLSQEDFLFMYGR VLATFGEKREIPNFDLVYQAMEAALVNRIGNETESKPLDVAKRILLSRGFGIRNTIDVDL LLKMQNEDGSWPL QVLSNLPSAKGGVFNSW DLSFAVRALQSQD

SEQ ID NO: 20

Artificial _SmHADl mutant of class II motif_E. coli optimized

ATGATCATTATCAGCTTCGCGTGCCTGCTGAAGTTTGGTTGCGGTGACAACGGTCCG CGTGGCGATCTGCTGC

GTCGTGCGCTGCAGCACAGCAGCTTCCTGGCGTATAGCTGCGGCGAGCTGGACCGTA CCGCGGCGATTAGCAC

CATCAGCCGTAAGTTTAAACTGACCGACCCGGCGCTGGTTGATAGCATGCTGCTGGA AGCGGCGAGCAGCTGC

GAGGTGGACAAAGAACTGCTGAGCCTGCTGCAGAGCACCCGTCAGCAACTGCTGGGT TGGATTGATATCCCGC

CGCAAGAGTGGGAACGTGTGTACAGCCTGTTCCCGGTTAGCCTGTGGAAGAACTTTG CGACCATTAGCCGTGA

CCTGGATAGCCTGCTGGGCGACATTCGTAGCCACGCGGTGATCGTTGATAAAAGCGT TGAGATGGCGGCGCTG

CACGCGTTCGAAAGCCTGCTGGCGCTGCCGTATGCGAGCCCGAACAAGAAAGCGTGC CAGGACTTCCTGAAGC

GTCGTTTTCTGCTGCCGCGTGCGGTGAAGCTGGGTATGGATCGTGTGAAAGAGGAAG TTCGTAGCAAGCAACT

GAAAAGCCTGTACCTGCTGGACAACGGCCGTCACGAGCTGGTTAGCGAAATTTTCTT TCCGTGCGTTGCGGCG

TGGTGCCTGCCGGAGATTATCCCGCTGGGTTGGATGAAGTATCTGCAGGTTCTGATT AAACGTGGCCACCCGT

TCGGTTACTTTGGTGCGGATGTGAGCCGTTATCCGCCGGCGATCGCGACCATGAGCA CCTGCGTGAGCACCCT

GTTCGATCTGAGCCTGGTTACCAGCGCGCAAGCGATGCACTTTCTGGAGATCTGCCT GGAAAACGTTAACGAC

CAGAACCAACTGCTGACCTACCTGGACGTGGAACGTCCGCGTGTTGATCCGGTGGTT ATTGCGAACGTGGTTT

ACCTGGCGTATGCGCTGAGCATGGAGGATCACCCGGTGGTTCGTCACAACGAAAACC TGATCCAGCGTTACCT

GCTGAGCGGTGGCTTCGTTTATGGCACCCGTTACTATCTGAGCCAAGAGGACTTCCT GTTTATGTACGGTCGT

GTGCTGGCGACCTTCGGCGAGAAACGTGAAATCCCGAACTTTGATCTGGTGTATCAG GCGATGGAAGCGGCGC

TGGTTAACCGTATTGGTAACGAGACCGAAAGCAAGCCGCTGGACGTTGCGAAACGTA TCCTGCTGAGCCGTGG

TTTTGGCATTCGTAACACCATCGACGTGGATCTGCTGCTGAAGATGCAGAACGAGGA TGGCAGCTGGCCGCTG

CAAGTGCTGAGCAACCTGCCGAGCGCGAAAGGTGGCGTTTTCAACAGCGTGGTTGAC CTGAGCTTTGCGGTGC

GTGCGCTGCAGAGCCAAGATTAA

SEQ ID NO: 21

Artificial _SmHADl mutant of class II motif

MIIISFACLLKFGCGDNGPRGDLLRRALQHSSFLAYSCGELDRTAAISTISRKFKLT DPALVDSMLLEAASSC EVDKELLSLLQSTRQQLLGWIDIPPQEWERVYSLFPVSLWKNFATISRDLDSLLGDIRSH AVIVDKSVEMAAL HAFESLLALPYASPNKKACQDFLKRRFLLPRAVKLGMDRVKEEVRSKQLKSLYLLDNGRH ELVSEIFFPCVAA WCLPEIIPLGWMKYLQVLIKRGHPFGYFGADVSRYPPAIATMSTCVSTLFDLSLVTSAQA MHFLEICLENVND QNQLLTYLDVERPRVDPW IANW YLAYALSMEDHPW RHNENLIQRYLLSGGFVYGTRYYLSQEDFLFMYGR VLATFGEKREIPNFDLVYQAMEAALVNRIGNETESKPLDVAKRILLSRGFGIRNTIDVDL LLKMQNEDGSWPL QVLSNLPSAKGGVFNSW DLSFAVRALQSQD

SEQ ID NO: 22

Artificial _SmHADl mutant of QW motif_E. coli optimized

ATGATCATTATCAGCTTCGCGTGCCTGCTGAAGTTTGGTTGCGGTGACAACGGTCCG CGTGGCGATCTGCTGC GTCGTGCGCTGCAGCACAGCAGCTTCCTGGCGTATAGCTGCGGCGAGCTGGACCGTACCG CGGCGATTAGCAC CATCAGCCGTAAGTTTAAACTGACCGACCCGGCGCTGGTTGATAGCATGCTGCTGGAAGC GGCGAGCAGCTGC GAGGTGGACAAAGAACTGCTGAGCCTGCTGCAGAGCACCCGTCAGCAACTGCTGGGTTGG ATTGATATCCCGC CGCAAGAGTGGGAACGTGTGTACAGCCTGTTCCCGGTTAGCCTGTGGAAGAACTTTGCGA CCATTAGCCGTGA CCTGGATAGCCTGCTGGGCGACATTCGTAGCCACGCGGTGATCGTTGATAAAAGCGTTGA GATGGCGGCGCTG CACGCGTTCGAAAGCCTGCTGGCGCTGCCGTATGCGAGCCCGAACAAGAAAGCGTGCCAG GACTTCCTGAAGC GTCGTTTTCTGCTGCCGCGTGCGGTGAAGCTGGGTATGGATCGTGTGAAAGAGGAAGTTC GTAGCAAGCAACT GAAAAGCCTGTACCTGCTGGACAACGGCCGTCACGAGCTGGTTAGCGAAATTTTCTTTCC GTGCGTTGCGGCG TGGTGCCTGCCGGAGATTATCCCGCTGGGTTGGATGAAGTATCTGCAGGTTCTGATTAAA CGTGGCCACCCGT TCGGTTACTTTGGTGCGGATGTGAGCCGTTATCCGCCGGACATCGATACCATGAGCACCT GCGTGAGCACCCT GTTCGATCTGAGCCTGGTTACCAGCGCGCAAGCGATGCACTTTCTGGAGATCTGCCTGGA AAACGTTAACGAC CAGAACCAACTGCTGACCTACCTGGACGTGGAACGTCCGCGTGTTGATCCGGTGGTTATT GCGAACGTGGTTT ACCTGGCGTATGCGCTGAGCATGGAGGATCACCCGGTGGTTCGTCACAACGAAAACCTGA TCCAGCGTTACCT GCTGAGCGGTGGCTTCGTTTATGGCACCCGTTACTATCTGAGCCAAGAGGACTTCCTGTT TATGTACGGTCGT GTGCTGGCGACCTTCGGCGAGAAACGTGAAATCCCGAACTTTGATCTGGTGTATCAGGCG ATGGAAGCGGCGC TGGTTAACCGTATTGGTAACGAGACCGAAAGCAAGCCGCTGGACGTTGCGAAACGTATCC TGCTGAGCCGTGG TTTTGGCATTCGTAACACCATCGACGTGGATCTGCTGCTGAAGATGCAGAACGCGGCGGG CAGCTGGCCGCTG CAAGTGCTGAGCAACCTGCCGAGCGCGAAAGGTGGCGTTTTCAACAGCGTGGTTGACCTG AGCTTTGCGGTGC GTGCGCTGCAGAGCCAAGATTAA

SEQ ID NO: 23

Artificial _SmHADl mutant of QW motif

MIIISFACLLKFGCGDNGPRGDLLRRALQHSSFLAYSCGELDRTAAISTISRKFKLT DPALVDSMLLEAASSC EVDKELLSLLQSTRQQLLGWIDIPPQEWERVYSLFPVSLWKNFATISRDLDSLLGDIRSH AVIVDKSVEMAAL HAFESLLALPYASPNKKACQDFLKRRFLLPRAVKLGMDRVKEEVRSKQLKSLYLLDNGRH ELVSEIFFPCVAA WCLPEIIPLGWMKYLQVLIKRGHPFGYFGADVSRYPPDIDTMSTCVSTLFDLSLVTSAQA MHFLEICLENVND QNQLLTYLDVERPRVDPW IANW YLAYALSMEDHPW RHNENLIQRYLLSGGFVYGTRYYLSQEDFLFMYGR VLATFGEKREIPNFDLVYQAMEAALVNRIGNETESKPLDVAKRILLSRGFGIRNTIDVDL LLKMQNAAGSWPL QVLSNLPSAKGGVFNSW DLSFAVRALQSQD

SEQ ID NO: 24

Selaginella moellendorffii_SmHAD2_wt_nucleic acid sequence

ATGATAATCATTAGTTTTGCTTGCTTGCTTAAGTTCGGATGTGGAGACAGTGGCCCC CGGGGAGATCTCCTCC GCAGGGCTCTCCAGCACTCCTCCTTCTTGGCCTATTCTTGCGGGGAGCTCGATCGCGCTG CCGCAATCTCGAC AATCTCGCGCAAATTCAAGCTCAAGGAGCCAGCTCTCCTGGATTCCATGCTGCTCGAGGC CGCTTCCAGCTGC GAGGTGGACGAGGAGCTACTGTCACTGCTCCAGTCAACGAGGCAGCAGCTCCTGGGATGG ATTGATATTCCTC CACAAGAGTGGGAGCGAGTTTACAACCTCTTCCCGTGGTCGCTCTGGAAGAACTTCGCTA CAATCTCCCGCGA TCTAGATTCCCTCCTTGGAGATATCAGGTTCCATGCTGTGATTGTCGATAAAAGCGTGGA GATGGCCGCGCTA CATGCCTTCGAGAGCTTGCTCGCTCTCCCGTATGCCTCGCCCAACAAGAAAGCTTGCCAG GACTTCTTGAAGC GTCGGTTCCTGCTGCCTCGAGCTGTGAAGCTGGGGATGGATCGAGTCAAGGAGGAGGTGA GATCCAAGCAGCT CAAATCACTCTACCTGCTTGACAACGGCCGGCAGGAGCTCGTCTCCGAGATCTTCTTTCC CTGCGTGGCGGCC TGGTGCCTTCCTGAGATCATTCCTCTCGGCTGGATGGAATCTCTCCAGGTACTGATCGAG CGCGGCCATCCCT TCGGTTACTTCGGTGCCGACGTCTCGCGCTATCCGCCGGACATCGACACCATGTCGACTT GTGTCTCCACACT TTTTGATCTTAGCCTCGTCACCTCCGCGCAAGCTATGCACTTTCTGGAGATTTGTCTCGA GAACGTCAACGAT CAAAACCAGCTTCTCACGTACCTGGACTTGGAGCGGCCTCGAGTGGATCCGGTCGTCATA GCCAACGTTGTCT ATTTCGCCTATGCGCTCTCCATGGAAGATCACCCGGTTGTCCGCCACAACGAGAATCTGA TCCAGAGATATCT GTTAAGCGGCGGATTCGTATACGGGACTCGCTATTACCTCAGCCAGGAGGACTTCTTGTT CATGTATGGCCGG GTCCTCGCGACGTTTGGAGAGAAGAGGGAGATTCCCAACTTTGATCTCGTGTATCAAGCG ATGGAGGCGGCTC TCGTCAACCGGATCGGGAACGAGACCGAGTCGAAGCCACTGGACGTTGCCAAAAGGATTC TGCTCAGCAGGTA CTTTGGAATCCGGAACACGATAGATGTGGACTTGCTCCTCAAGATGCAGAACGAGGATGG TTCCTGGCCCTTG CAAGTGCTCAGCAATCTTCCATCTGCTAAAGGTGGTGTCTTCAATTCGGTGGTGGACTTG AGCTTTGCGGTGA GAGCACTGCAAAGTCAAGATTGA

SEQ ID NO: 25

Selaginella moellendorffli_SmHAO2_E. coll optimized

ATGATCATTATCAGCTTCGCGTGCCTGCTGAAATTTGGTTGCGGTGATAGCGGTCCG CGTGGCGATCTGCTGC GTCGTGCGCTGCAGCACAGCAGCTTCCTGGCGTATAGCTGCGGCGAGCTGGACCGTGCGG CGGCGATTAGCAC CATCAGCCGTAAGTTTAAACTGAAGGAACCGGCGCTGCTGGACAGCATGCTGCTGGAGGC GGCGAGCAGCTGC GAAGTTGATGAGGAACTGCTGAGCCTGCTGCAGAGCACCCGTCAGCAACTGCTGGGTTGG ATTGATATCCCGC CGCAAGAGTGGGAACGTGTGTATAACCTGTTCCCGTGGAGCCTGTGGAAAAACTTTGCGA CCATTAGCCGTGA CCTGGATAGCCTGCTGGGCGACATTCGTTTCCACGCGGTGATCGTTGATAAGAGCGTTGA GATGGCGGCGCTG CACGCGTTTGAAAGCCTGCTGGCGCTGCCGTACGCGAGCCCGAACAAGAAAGCGTGCCAA GACTTCCTGAAGC GTCGTTTTCTGCTGCCGCGTGCGGTGAAACTGGGTATGGATCGTGTGAAGGAAGAGGTGC GCAGCAAACAGCT GAAGAGCCTGTATCTGCTGGACAACGGCCGTCAAGAGCTGGTTAGCGAAATTTTCTTTCC GTGCGTTGCGGCG TGGTGCCTGCCGGAGATTATCCCGCTGGGTTGGATGGAGAGCCTGCAGGTTCTGATTGAA CGTGGCCACCCGT TCGGTTACTTTGGTGCGGATGTGAGCCGTTATCCGCCGGACATCGATACCATGAGCACCT GCGTGAGCACCCT GTTCGATCTGAGCCTGGTTACCAGCGCGCAAGCGATGCACTTTCTGGAGATCTGCCTGGA AAACGTTAACGAC CAGAACCAACTGCTGACCTACCTGGACCTGGAACGTCCGCGTGTGGATCCGGTGGTTATT GCGAACGTGGTTT ACTTCGCGTATGCGCTGAGCATGGAGGATCACCCGGTGGTTCGTCACAACGAAAACCTGA TCCAGCGTTACCT GCTGAGCGGTGGCTTTGTTTATGGCACCCGTTACTATCTGAGCCAAGAGGACTTCCTGTT TATGTACGGTCGT GTGCTGGCGACCTTCGGCGAGAAACGTGAAATCCCGAACTTTGATCTGGTGTATCAGGCG ATGGAAGCGGCGC TGGTTAACCGTATTGGCAACGAGACCGAAAGCAAACCGCTGGACGTTGCGAAGCGTATCC TGCTGAGCCGTTA CTTCGGTATTCGTAACACCATCGACGTGGATCTGCTGCTGAAAATGCAGAACGAGGATGG CAGCTGGCCGCTG CAAGTGCTGAGCAACCTGCCGAGCGCGAAGGGTGGCGTTTTCAACAGCGTGGTTGACCTG AGCTTTGCGGTGC GTGCGCTGCAGAGCCAAGATTAA

SEQ ID NO: 26

Selaginella moellendorffii _SmHAD2_amino acid sequence

MIIISFACLLKFGCGDSGPRGDLLRRALQHSSFLAYSCGELDRAAAISTISRKFKLK EPALLDSMLLEAASSC EVDEELLSLLQSTRQQLLGWIDIPPQEWERVYNLFPWSLWKNFATISRDLDSLLGDIRFH AVIVDKSVEMAAL HAFESLLALPYASPNKKACQDFLKRRFLLPRAVKLGMDRVKEEVRSKQLKSLYLLDNGRQ ELVSEIFFPCVAA WCLPEIIPLGWMESLQVLIERGHPFGYFGADVSRYPPDIDTMSTCVSTLFDLSLVTSAQA MHFLEICLENVND QNQLLTYLDLERPRVDPW IANW YFAYALSMEDHPW RHNENLIQRYLLSGGFVYGTRYYLSQEDFLFMYGR VLATFGEKREIPNFDLVYQAMEAALVNRIGNETESKPLDVAKRILLSRYFGIRNTIDVDL LLKMQNEDGSWPL QVLSNLPSAKGGVFNSW DLSFAVRALQSQD

SEQ ID NO: 27

Artificial _SmHAD2 mutant of class II motif_E. coli optimized

ATGATCATTATCAGCTTCGCGTGCCTGCTGAAATTTGGTTGCGGTGATAGCGGTCCG CGTGGCGATCTGCTGC GTCGTGCGCTGCAGCACAGCAGCTTCCTGGCGTATAGCTGCGGCGAGCTGGACCGTGCGG CGGCGATTAGCAC CATCAGCCGTAAGTTTAAACTGAAGGAACCGGCGCTGCTGGACAGCATGCTGCTGGAGGC GGCGAGCAGCTGC GAAGTTGATGAGGAACTGCTGAGCCTGCTGCAGAGCACCCGTCAGCAACTGCTGGGTTGG ATTGATATCCCGC CGCAAGAGTGGGAACGTGTGTATAACCTGTTCCCGTGGAGCCTGTGGAAAAACTTTGCGA CCATTAGCCGTGA CCTGGATAGCCTGCTGGGCGACATTCGTTTCCACGCGGTGATCGTTGATAAGAGCGTTGA GATGGCGGCGCTG CACGCGTTTGAAAGCCTGCTGGCGCTGCCGTACGCGAGCCCGAACAAGAAAGCGTGCCAA GACTTCCTGAAGC GTCGTTTTCTGCTGCCGCGTGCGGTGAAACTGGGTATGGATCGTGTGAAGGAAGAGGTGC GCAGCAAACAGCT GAAGAGCCTGTATCTGCTGGACAACGGCCGTCAAGAGCTGGTTAGCGAAATTTTCTTTCC GTGCGTTGCGGCG TGGTGCCTGCCGGAGATTATCCCGCTGGGTTGGATGGAGAGCCTGCAGGTTCTGATTGAA CGTGGCCACCCGT TCGGTTACTTTGGTGCGGATGTGAGCCGTTATCCGCCGGCGATCGCGACCATGAGCACCT GCGTGAGCACCCT GTTCGATCTGAGCCTGGTTACCAGCGCGCAAGCGATGCACTTTCTGGAGATCTGCCTGGA AAACGTTAACGAC CAGAACCAACTGCTGACCTACCTGGACCTGGAACGTCCGCGTGTGGATCCGGTGGTTATT GCGAACGTGGTTT ACTTCGCGTATGCGCTGAGCATGGAGGATCACCCGGTGGTTCGTCACAACGAAAACCTGA TCCAGCGTTACCT GCTGAGCGGTGGCTTTGTTTATGGCACCCGTTACTATCTGAGCCAAGAGGACTTCCTGTT TATGTACGGTCGT GTGCTGGCGACCTTCGGCGAGAAACGTGAAATCCCGAACTTTGATCTGGTGTATCAGGCG ATGGAAGCGGCGC TGGTTAACCGTATTGGCAACGAGACCGAAAGCAAACCGCTGGACGTTGCGAAGCGTATCC TGCTGAGCCGTTA CTTCGGTATTCGTAACACCATCGACGTGGATCTGCTGCTGAAAATGCAGAACGAGGATGG CAGCTGGCCGCTG CAAGTGCTGAGCAACCTGCCGAGCGCGAAGGGTGGCGTTTTCAACAGCGTGGTTGACCTG AGCTTTGCGGTGC GTGCGCTGCAGAGCCAAGATTAA

SEQ ID NO: 28

Artificial_SmHAD2 mutant of class II motif

MIIISFACLLKFGCGDSGPRGDLLRRALQHSSFLAYSCGELDRAAAISTISRKFKLK EPALLDSMLLEAASSC EVDEELLSLLQSTRQQLLGWIDIPPQEWERVYNLFPWSLWKNFATISRDLDSLLGDIRFH AVIVDKSVEMAAL HAFESLLALPYASPNKKACQDFLKRRFLLPRAVKLGMDRVKEEVRSKQLKSLYLLDNGRQ ELVSEIFFPCVAA WCLPEIIPLGWMESLQVLIERGHPFGYFGADVSRYPPAIATMSTCVSTLFDLSLVTSAQA MHFLEICLENVND QNQLLTYLDLERPRVDPW IANW YFAYALSMEDHPW RHNENLIQRYLLSGGFVYGTRYYLSQEDFLFMYGR VLATFGEKREIPNFDLVYQAMEAALVNRIGNETESKPLDVAKRILLSRYFGIRNTIDVDL LLKMQNEDGSWPL QVLSNLPSAKGGVFNSW DLSFAVRALQSQD

SEQ ID NO: 29

Artificial _SmHAD2 mutant of QW motif_E. coli optimized

ATGATCATTATCAGCTTCGCGTGCCTGCTGAAATTTGGTTGCGGTGATAGCGGTCCG CGTGGCGATCTGCTGC

GTCGTGCGCTGCAGCACAGCAGCTTCCTGGCGTATAGCTGCGGCGAGCTGGACCGTG CGGCGGCGATTAGCAC CATCAGCCGTAAGTTTAAACTGAAGGAACCGGCGCTGCTGGACAGCATGCTGCTGGAGGC GGCGAGCAGCTGC GAAGTTGATGAGGAACTGCTGAGCCTGCTGCAGAGCACCCGTCAGCAACTGCTGGGTTGG ATTGATATCCCGC CGCAAGAGTGGGAACGTGTGTATAACCTGTTCCCGTGGAGCCTGTGGAAAAACTTTGCGA CCATTAGCCGTGA CCTGGATAGCCTGCTGGGCGACATTCGTTTCCACGCGGTGATCGTTGATAAGAGCGTTGA GATGGCGGCGCTG CACGCGTTTGAAAGCCTGCTGGCGCTGCCGTACGCGAGCCCGAACAAGAAAGCGTGCCAA GACTTCCTGAAGC GTCGTTTTCTGCTGCCGCGTGCGGTGAAACTGGGTATGGATCGTGTGAAGGAAGAGGTGC GCAGCAAACAGCT GAAGAGCCTGTATCTGCTGGACAACGGCCGTCAAGAGCTGGTTAGCGAAATTTTCTTTCC GTGCGTTGCGGCG TGGTGCCTGCCGGAGATTATCCCGCTGGGTTGGATGGAGAGCCTGCAGGTTCTGATTGAA CGTGGCCACCCGT TCGGTTACTTTGGTGCGGATGTGAGCCGTTATCCGCCGGACATCGATACCATGAGCACCT GCGTGAGCACCCT GTTCGATCTGAGCCTGGTTACCAGCGCGCAAGCGATGCACTTTCTGGAGATCTGCCTGGA AAACGTTAACGAC CAGAACCAACTGCTGACCTACCTGGACCTGGAACGTCCGCGTGTGGATCCGGTGGTTATT GCGAACGTGGTTT ACTTCGCGTATGCGCTGAGCATGGAGGATCACCCGGTGGTTCGTCACAACGAAAACCTGA TCCAGCGTTACCT GCTGAGCGGTGGCTTTGTTTATGGCACCCGTTACTATCTGAGCCAAGAGGACTTCCTGTT TATGTACGGTCGT GTGCTGGCGACCTTCGGCGAGAAACGTGAAATCCCGAACTTTGATCTGGTGTATCAGGCG ATGGAAGCGGCGC TGGTTAACCGTATTGGCAACGAGACCGAAAGCAAACCGCTGGACGTTGCGAAGCGTATCC TGCTGAGCCGTTA CTTCGGTATTCGTAACACCATCGACGTGGATCTGCTGCTGAAAATGCAGAACGCGGCGGG CAGCTGGCCGCTG CAAGTGCTGAGCAACCTGCCGAGCGCGAAGGGTGGCGTTTTCAACAGCGTGGTTGACCTG AGCTTTGCGGTGC GTGCGCTGCAGAGCCAAGATTAA

SEQ ID NO: 30

Artificial _SmHAD2 mutant of QW motif

MIIISFACLLKFGCGDSGPRGDLLRRALQHSSFLAYSCGELDRAAAISTISRKFKLK EPALLDSMLLEAASSC EVDEELLSLLQSTRQQLLGWIDIPPQEWERVYNLFPWSLWKNFATISRDLDSLLGDIRFH AVIVDKSVEMAAL HAFESLLALPYASPNKKACQDFLKRRFLLPRAVKLGMDRVKEEVRSKQLKSLYLLDNGRQ ELVSEIFFPCVAA WCLPEIIPLGWMESLQVLIERGHPFGYFGADVSRYPPDIDTMSTCVSTLFDLSLVTSAQA MHFLEICLENVND QNQLLTYLDLERPRVDPW IANW YFAYALSMEDHPW RHNENLIQRYLLSGGFVYGTRYYLSQEDFLFMYGR VLATFGEKREIPNFDLVYQAMEAALVNRIGNETESKPLDVAKRILLSRYFGIRNTIDVDL LLKMQNAAGSWPL QVLSNLPSAKGGVFNSW DLSFAVRALQSQD

SEQ ID NO: 31

Gelatoporia subvermispora _EMD37666.1_E. coli optimized

ATGTCTGCGGCGGCTCAATACACGACTTTGATTCTGGATCTGGGTGATGTTCTGTTC ACTTGGTCCCCGAAAA CCAAGACCAGCATCCCTCCGCGTACCCTGAAAGAAATCCTGAATAGCGCTACCTGGTATG AGTACGAGCGTGG TCGCATTTCCCAAGACGAGTGTTACGAACGTGTGGGCACCGAGTTCGGCATTGCGCCGAG CGAGATTGACAAC GCGTTCAAACAAGCGCGCGATTCGATGGAAAGCAATGATGAACTGATCGCACTGGTCCGT GAGCTGAAAACGC AGCTGGACGGTGAGCTGCTGGTTTTCGCACTGTCCAATATTAGCCTGCCGGATTACGAAT ACGTCTTGACCAA ACCGGCGGACTGGAGCATCTTTGACAAAGTGTTCCCTAGCGCCTTGGTGGGCGAGCGTAA GCCGCATCTGGGC GTTTATAAACACGTTATTGCGGAAACGGGCATTGATCCGCGCACGACGGTTTTCGTGGAC GACAAGATTGACA ATGTGTTAAGCGCACGCAGCGTCGGTATGCATGGTATCGTGTTTGAGAAACAAGAAGATG TCATGCGTGCACT GCGTAACATCTTTGGTGATCCGGTCCGTCGTGGTCGTGAGTATCTGCGTAGAAACGCAAT GCGTCTGGAGTCC GTGACCGACCACGGCGTGGCGTTTGGTGAGAACTTTACCCAGTTGCTGATTCTGGAATTG ACGAACGACCCGA GCCTGGTCACCCTGCCTGATCGTCCGCGTACCTGGAACTTTTTTCGCGGCAATGGTGGCC GCCCGAGCAAGCC GCTGTTCAGCGAAGCGTTCCCGGATGATCTGGATACCACGAGCCTGGCGCTGACCGTGCT GCAGCGCGACCCG GGTGTTATCAGCAGCGTTATGGACGAAATGCTGAATTACCGTGACCCGGACGGTATCATG CAGACTTATTTCG ATGACGGTCGCCAACGCTTGGACCCATTTGTGAACGTCAATGTTCTGACCTTTTTCTATA CGAACGGCCGTGG TCACGAACTGGACCAGTGTCTGACGTGGGTGCGTGAAGTCCTCTTGTATCGTGCGTACCT TGGTGGCTCACGC TACTACCCATCGGCGGATTGCTTCCTGTACTTCATCTCTCGTCTGTTTGCGTGTACCAAT GACCCGGTGCTGC ACCATCAGCTGAAGCCACTGTTTGTTGAGCGTGTCCAAGAGCAAATTGGTGTCGAGGGTG ATGCACTGGAACT GGCTTTTCGTCTGCTGGTCTGCGCCAGCCTGGATGTCCAGAATGCCATCGACATGCGCCG TCTGCTGGAAATG CAGTGCGAAGATGGCGGTTGGGAGGGTGGTAACCTCTACCGCTTCGGCACCACGGGCCTG AAAGTTACCAACC GCGGTCTGACGACCGCAGCCGCCGTTCAAGCGATCGAAGCGAGCCAACGCCGTCCGCCGA GCCCGAGCCCGTC TGTAGAGAGCACGAAAAGCCCGATTACCCCGGTGACCCCGATGCTGGAAGTTCCAAGCCT GGGCTTATCTATC AGCCGTCCGTCCAGCCCGCTGCTGGGTTATTTCCGTTTGCCGTGGAAGAAAAGCGCAGAA GTGCACTAA

SEQ ID NO: 32

Gelatoporia subvermispora _EMD37666.l_amino acid sequence

MSAAAQYTTLILDLGDVLFTWSPKTKTSIPPRTLKEILNSATWYEYERGRISQDECY ERVGTEFGIAPSEIDN AFKQARDSMESNDELIALVRELKTQLDGELLVFALSNISLPDYEYVLTKPADWSIFDKVF PSALVGERKPHLG VYKHVIAETGIDPRTTVFVDDKIDNVLSARSVGMHGIVFEKQEDVMRALRNIFGDPVRRG REYLRRNAMRLES VTDHGVAFGENFTQLLILELTNDPSLVTLPDRPRTWNFFRGNGGRPSKPLFSEAFPDDLD TTSLALTVLQRDP GVISSVMDEMLNYRDPDGIMQTYFDDGRQRLDPFVNVNVLTFFYTNGRGHELDQCLTWVR EVLLYRAYLGGSR YYPSADCFLYFISRLFACTNDPVLHHQLKPLFVERVQEQIGVEGDALELAFRLLVCASLD VQNAIDMRRLLEM QCEDGGWEGGNLYRFGTTGLKVTNRGLTTAAAVQAIEASQRRPPSPSPSVESTKSPITPV TPMLEVPSLGLSI SRPSSPLLGYFRLPWKKSAEVH

SEQ ID NO: 33

Artificial _EMD37666.l_mutant of class II motif_E.coli optimized

ATGAGCGCGGCGGCGCAGTACACCACCCTGATCCTGGACCTGGGTGATGTGCTGTTC ACCTGGAGCCCGAAGA CCAAAACCAGCATCCCGCCGCGTACCCTGAAGGAAATTCTGAACAGCGCGACCTGGTACG AGTATGAACGTGG CCGTATCAGCCAAGACGAGTGCTATGAACGTGTTGGCACCGAGTTCGGCATCGCGCCGAG CGAAATTGATAAC GCGTTTAAGCAGGCGCGTGACAGCATGGAGAGCAACGATGAACTGATTGCGCTGGTGCGT GAGCTGAAAACCC AACTGGACGGTGAACTGCTGGTTTTCGCGCTGAGCAACATCAGCCTGCCGGATTACGAGT ATGTGCTGACCAA GCCGGCGGACTGGAGCATTTTCGATAAAGTTTTTCCGAGCGCGCTGGTTGGTGAACGCAA GCCGCACCTGGGC GTTTACAAACACGTGATCGCGGAAACCGGTATTGACCCGCGTACCACCGTGTTTGTTGAC GATAAGATCGATA ACGTTCTGAGCGCGCGTAGCGTGGGTATGCACGGCATTGTTTTCGAGAAACAGGAAGACG TGATGCGTGCGCT GCGTAACATCTTTGGTGATCCGGTTCGTCGTGGCCGTGAGTATCTGCGTCGTAACGCGAT GCGTCTGGAAAGC GTTACCGACCACGGTGTGGCGTTCGGCGAGAACTTTACCCAACTGCTGATTCTGGAACTG ACCAACGATCCGA GCCTGGTGACCCTGCCGGATCGTCCGCGTACCTGGAACTTCTTTCGTGGTAACGGTGGCC GTCCGAGCAAGCC GCTGTTCAGCGAAGCGTTTCCGGCGGCGCTGGCGACCACCAGCCTGGCGCTGACCGTTCT GCAGCGTGACCCG GGCGTTATCAGCAGCGTGATGGATGAGATGCTGAACTACCGTGACCCGGATGGTATTATG CAGACCTATTTCG ACGATGGCCGTCAACGTCTGGACCCGTTTGTGAACGTTAACGTGCTGACCTTCTTTTACA CCAACGGTCGTGG CCACGAGCTGGATCAGTGCCTGACCTGGGTTCGTGAAGTGCTGCTGTACCGTGCGTATCT GGGTGGCAGCCGT TACTATCCGAGCGCGGACTGCTTCCTGTATTTTATCAGCCGTCTGTTCGCGTGCACCAAC GATCCGGTGCTGC ACCACCAACTGAAACCGCTGTTTGTTGAGCGTGTGCAGGAACAAATCGGTGTTGAGGGCG ACGCGCTGGAACT GGCGTTCCGTCTGCTGGTGTGCGCGAGCCTGGACGTTCAGAACGCGATTGATATGCGTCG TCTGCTGGAGATG CAATGCGAAGATGGTGGCTGGGAAGGTGGCAACCTGTACCGTTTTGGCACCACCGGCCTG AAGGTGACCAACC GTGGTCTGACCACCGCGGCGGCGGTTCAGGCGATTGAGGCGAGCCAACGTCGTCCGCCGA GCCCGAGCCCGAG CGTGGAAAGCACCAAAAGCCCGATCACCCCGGTGACCCCGATGCTGGAAGTGCCGAGCCT GGGTCTGAGCATT AGCCGTCCGAGCAGCCCGCTGCTGGGTTACTTCCGTCTGCCGTGGAAGAAAAGCGCGGAA GTGCACTAA

SEQ ID NO: 34

Artificial _EMD37666.l_mutant of class II motif

MSAAAQYTTLILDLGDVLFTWSPKTKTSIPPRTLKEILNSATWYEYERGRISQDECY ERVGTEFGIAPSEIDN AFKQARDSMESNDELIALVRELKTQLDGELLVFALSNISLPDYEYVLTKPADWSIFDKVF PSALVGERKPHLG VYKHVIAETGIDPRTTVFVDDKIDNVLSARSVGMHGIVFEKQEDVMRALRNIFGDPVRRG REYLRRNAMRLES VTDHGVAFGENFTQLLILELTNDPSLVTLPDRPRTWNFFRGNGGRPSKPLFSEAFPAALA TTSLALTVLQRDP GVISSVMDEMLNYRDPDGIMQTYFDDGRQRLDPFVNVNVLTFFYTNGRGHELDQCLTWVR EVLLYRAYLGGSR YYPSADCFLYFISRLFACTNDPVLHHQLKPLFVERVQEQIGVEGDALELAFRLLVCASLD VQNAIDMRRLLEM QCEDGGWEGGNLYRFGTTGLKVTNRGLTTAAAVQAIEASQRRPPSPSPSVESTKSPITPV TPMLEVPSLGLSI SRPSSPLLGYFRLPWKKSAEVH coli optimized

ATGGCGAGCATCCACCGTCGTTACACCACCCTGATTCTGGACCTGGGTGATGTTCTG TTTCGTTGGAGCCCGA AGACCGAGACCGCGATCCCGCCGCAGCAACTGAAAGACATTCTGAGCAGCGTGACCTGGT TCGAGTACGAACG TGGCCGTCTGAGCCAGGAAGCGTGCTATGAACGTTGCGCGGAGGAATTTAAGATCGAGGC GAGCGTTATTGCG GAAGCGTTCAAACAAGCGCGTGGTAGCCTGCGTCCGAACGAGGAATTTATCGCGCTGATT CGTGACCTGCGTC GTGAGATGCACGGCGATCTGACCGTGCTGGCGCTGAGCAACATCAGCCTGCCGGACTACG AATATATTATGAG CCTGAGCAGCGACTGGACCACCGTTTTCGATCGTGTGTTTCCGAGCGCGCTGGTTGGTGA GCGTAAGCCGCAC CTGGGCTGCTATCGTAAAGTGATCAGCGAGATGAACCTGGAACCGCAGACCACCGTGTTC GTTGACGATAAGC TGGATAACGTTGCGAGCGCGCGTAGCCTGGGTATGCATGGTATTGTTTTCGACAACCAGG CGAACGTGTTTCG TCAACTGCGTAACATCTTCGGTGATCCGATTCGTCGTGGCCAAGAGTACCTGCGTGGTCA CGCGGGCAAACTG GAAAGCAGCACCGACAACGGTCTGATCTTCGAGGAAAACTTTACCCAGCTGATCATTTAT GAGCTGACCCAAG ATCGTACCCTGATTAGCCTGAGCGAATGCCCGCGTACCTGGAACTTCTTTCGTGGCGAGC CGCTGTTCAGCGA AACCTTCCCGGACGACGTTGACACCACCAGCGTTGCGCTGACCGTGCTGCAGCCGGACCG TGCGCTGGTTAAC AGCGTGCTGGATGAGATGCTGGAATACGTGGACGCGGATGGTATCATGCAAACCTATTTT GACCGTAGCCGTC CGCGTATGGATCCGTTTGTGTGCGTTAACGTGCTGAGCCTGTTCTACGAGAACGGTCGTG GCCACGAACTGCC

GCGTACCCTGGACTGGGTTTACGAGGTGCTGCTGCACCGTGCGTATCACGGTGGCAG CCGTTACTATCTGAGC

CCGGATTGCTTCCTGTTCTTTATGAGCCGTCTGCTGAAACGTGCGGATGATCCGGCG GTTCAGGCGCGTCTGC

GTCCGCTGTTTGTTGAGCGTGTGAACGAACGTGTTGGTGCGGCGGGTGACAGCATGG ATCTGGCGTTCCGTAT

CCTGGCGGCGGCGAGCGTTGGTGTGCAGTGCCCGCGTGACCTGGAACGTCTGACCGC GGGTCAATGCGATGAT

GGTGGCTGGGATCTGTGCTGGTTCTACGTTTTTGGTAGCACCGGCGTGAAAGCGGGT AACCGTGGCCTGACCA

CCGCGCTGGCGGTGACCGCGATTCAAACCGCGATTGGTCGTCCGCCGAGCCCGAGCC CGAGCGCGGCGAGCAG

CAGCTTTCGTCCGAGCAGCCCGTATAAATTCCTGGGTATCAGCCGTCCGGCGAGCCC GATTCGTTTTGGTGAC

CTGCTGCGTCCGTGGCGTAAGATGAGCCGTAGCAACCTGAAAAGCCAATAA acid sequence

MASIHRRYTTLILDLGDVLFRWSPKTETAIPPQQLKDILSSVTWFEYERGRLSQEAC YERCAEEFKIEASVIA EAFKQARGSLRPNEEFIALIRDLRREMHGDLTVLALSNISLPDYEYIMSLSSDWTTVFDR VFPSALVGERKPH LGCYRKVISEMNLEPQTTVFVDDKLDNVASARSLGMHGIVFDNQANVFRQLRNIFGDPIR RGQEYLRGHAGKL ESSTDNGLIFEENFTQLIIYELTQDRTLISLSECPRTWNFFRGEPLFSETFPDDVDTTSV ALTVLQPDRALVN SVLDEMLEYVDADGIMQTYFDRSRPRMDPFVCVNVLSLFYENGRGHELPRTLDWVYEVLL HRAYHGGSRYYLS PDCFLFFMSRLLKRADDPAVQARLRPLFVERVNERVGAAGDSMDLAFRILAAASVGVQCP RDLERLTAGQCDD GGWDLCWFYVFGSTGVKAGNRGLTTALAVTAIQTAIGRPPSPSPSAASSSFRPSSPYKFL GISRPASPIRFGD LLRPWRKMSRSNLKSQ

SEQ ID NO: 37

Artificial _XP_007369631.l_mutant of class II motif_ E.coli optimized

ATGGCGAGCATCCACCGTCGTTACACCACCCTGATTCTGGACCTGGGTGATGTTCTG TTTCGTTGGAGCCCGA AGACCGAGACCGCGATCCCGCCGCAGCAACTGAAAGACATTCTGAGCAGCGTGACCTGGT TCGAGTACGAACG TGGCCGTCTGAGCCAGGAAGCGTGCTATGAACGTTGCGCGGAGGAATTTAAGATCGAGGC GAGCGTTATTGCG GAAGCGTTCAAACAAGCGCGTGGTAGCCTGCGTCCGAACGAGGAATTTATCGCGCTGATT CGTGACCTGCGTC GTGAGATGCACGGCGATCTGACCGTGCTGGCGCTGAGCAACATCAGCCTGCCGGACTACG AATATATTATGAG CCTGAGCAGCGACTGGACCACCGTTTTCGATCGTGTGTTTCCGAGCGCGCTGGTTGGTGA GCGTAAGCCGCAC CTGGGCTGCTATCGTAAAGTGATCAGCGAGATGAACCTGGAACCGCAGACCACCGTGTTC GTTGACGATAAGC TGGATAACGTTGCGAGCGCGCGTAGCCTGGGTATGCATGGTATTGTTTTCGACAACCAGG CGAACGTGTTTCG TCAACTGCGTAACATCTTCGGTGATCCGATTCGTCGTGGCCAAGAGTACCTGCGTGGTCA CGCGGGCAAACTG GAAAGCAGCACCGACAACGGTCTGATCTTCGAGGAAAACTTTACCCAGCTGATCATTTAT GAGCTGACCCAAG ATCGTACCCTGATTAGCCTGAGCGAATGCCCGCGTACCTGGAACTTCTTTCGTGGCGAGC CGCTGTTCAGCGA AACCTTCCCGGCGGCGGTTGCGACCACCAGCGTTGCGCTGACCGTGCTGCAGCCGGACCG TGCGCTGGTTAAC AGCGTGCTGGATGAGATGCTGGAATACGTGGACGCGGATGGTATCATGCAAACCTATTTT GACCGTAGCCGTC CGCGTATGGATCCGTTTGTGTGCGTTAACGTGCTGAGCCTGTTCTACGAGAACGGTCGTG GCCACGAACTGCC GCGTACCCTGGACTGGGTTTACGAGGTGCTGCTGCACCGTGCGTATCACGGTGGCAGCCG TTACTATCTGAGC CCGGATTGCTTCCTGTTCTTTATGAGCCGTCTGCTGAAACGTGCGGATGATCCGGCGGTT CAGGCGCGTCTGC GTCCGCTGTTTGTTGAGCGTGTGAACGAACGTGTTGGTGCGGCGGGTGACAGCATGGATC TGGCGTTCCGTAT CCTGGCGGCGGCGAGCGTTGGTGTGCAGTGCCCGCGTGACCTGGAACGTCTGACCGCGGG TCAATGCGATGAT GGTGGCTGGGATCTGTGCTGGTTCTACGTTTTTGGTAGCACCGGCGTGAAAGCGGGTAAC CGTGGCCTGACCA CCGCGCTGGCGGTGACCGCGATTCAAACCGCGATTGGTCGTCCGCCGAGCCCGAGCCCGA GCGCGGCGAGCAG CAGCTTTCGTCCGAGCAGCCCGTATAAATTCCTGGGTATCAGCCGTCCGGCGAGCCCGAT TCGTTTTGGTGAC CTGCTGCGTCCGTGGCGTAAGATGAGCCGTAGCAACCTGAAAAGCCAATAA

SEQ ID NO: 38

Artificial_XP_007369631.l_mutant of class II motif

MASIHRRYTTLILDLGDVLFRWSPKTETAIPPQQLKDILSSVTWFEYERGRLSQEAC YERCAEEFKIEASVIA EAFKQARGSLRPNEEFIALIRDLRREMHGDLTVLALSNISLPDYEYIMSLSSDWTTVFDR VFPSALVGERKPH LGCYRKVISEMNLEPQTTVFVDDKLDNVASARSLGMHGIVFDNQANVFRQLRNIFGDPIR RGQEYLRGHAGKL ESSTDNGLIFEENFTQLIIYELTQDRTLISLSECPRTWNFFRGEPLFSETFPAAVATTSV ALTVLQPDRALVN SVLDEMLEYVDADGIMQTYFDRSRPRMDPFVCVNVLSLFYENGRGHELPRTLDWVYEVLL HRAYHGGSRYYLS PDCFLFFMSRLLKRADDPAVQARLRPLFVERVNERVGAAGDSMDLAFRILAAASVGVQCP RDLERLTAGQCDD GGWDLCWFYVFGSTGVKAGNRGLTTALAVTAIQTAIGRPPSPSPSAASSSFRPSSPYKFL GISRPASPIRFGD LLRPWRKMSRSNLKSQ

SEQ ID NO: 39 Artificial_class II synthase motif_mutated_BtHAD PDALASTS

SEQ ID NO: 40

Artificial_class II synthase motif_mutated_SmHADl+2

PPAIATMS SEQ ID NO: 41

Artificial_class II synthase motif_mutated_EMD37666 PDAALATTS

SEQ ID NO: 42

Artificial_class II synthase motif_mutated_XP_007369631

PDAAVATTS

SEQ ID NO: 43

Artificial_QW motif_mutated_BtHAD QNVAGSW

SEQ ID NO: 44

Artificia1_QW motif_mutated_SmHAD1+2 QNAAGSW

SEQ ID NO: 45

Artificial_class I synthase motif DDxx(D/E)

DDxx(D/E)

SEQ ID NO: 46

Artificial_class II synthase motif PxDxD(T/S)(T/M)S PxDxD(T/S)(T/M)S

SEQ I

Artif synthase motif PDDLDSTS PDDLD

SEQ ID NO: 48

Artificial_class II synthase motif PDDLDTTS

PDDLDTTS SEQ ID NO: 49

Artificial_class II synthase motif PPDIDTMS

PPDIDTMS

SEQ ID NO: 50

Artificial_class II synthase motif PNDIDTMS PNDIDTTS

SEQ ID NO: 51 Artificial_QW motif

QxxDGxW

SEQ ID NO: 52

Artificial_QW motif QNVDGSW QNVDGSW

SEQ ID NO: 53

Artificial_QW motif QCDDGGW QCDDGGW

SEQ ID NO: 54

Artificial_QW motif QSSDGGW QSSDGGW

SEQ ID NO: 55

Artificial_QW motif QNEDGSW QNEDGSW

SEQ ID NO: 56

Artificial_conserved motif 1 Lxxxx(W/F)xxYxxG Lxxxx(W/F)xxYxxG

SEQ ID NO: 57

Artificial_conserved motif 1 LRSHIWFNYSMG LRSHIWFNYSMG

SEQ ID NO: 58

Artificial_conserved motif 1 LRSATWAAYECG LRSATWAAYECG

SEQ ID NO: 59

Artificial_conserved motif 1 LRTPTWGKYECG LRTPTWGKYECG

SEQ ID NO: 60

Artificial_conserved motif 1 LQHSSFLAYSCG LQHSSFLAYSCG

SEQ ID NO: 61

Artificial_conserved motif 1 LRxxTWxxYECG

LRxxTWxxYECG SEQ ID NO: 62

Artificial_conserved motif 2 YxDxxRxRVD(P/A)V(V/A)xxN YxDxxRxRVDxVxxxN

SEQ ID NO: 63

Artificial_conserved motif 2 YFDPLRLRVDPVAATN YFDPLRLRVDPVAATN

SEQ ID NO: 64

Artificial_conserved motif 2 YFDETRPRVDAWNVN YFDETRPRVDAWNVN

SEQ ID NO: 65

Artificial_conserved motif 2 YFDKTRPRVDPWCVN

YFDKTRPRVDPWCVN SEQ ID NO: 66

Artificial_conserved motif 2 YLDVERPRVDPW IAN YLDVERPRVDPW IAN

SEQ ID NO: 67

Artificial_conserved motif 2 YLDLERPRVDPW IAN YLDLERPRVDPW IAN

SEQ ID NO: 68

Artificial_conserved motif 2 Y(F/L)Dx(T/E)RPRVD(P/A)W x(A/V)N YxDxxRPRVDxW xxN

SEQ ID NO: 69

Artificial_conserved motif 3 GTx(Y/F)YxxxExFL(Y/F)

GTxxYxxxExFLx

SEQ ID NO: 70

Artificial_conserved motif 3 GTLYYRTPEAFLY GTLYYRTPEAFLY

SEQ ID NO: 71

Artificial_conserved motif 3 GTLFYYHAESFLY GTLFYYHAESFLY

SEQ ID NO: 72

Artificial_conserved motif 3 GTRYYLSQEDFLF GTRYYLSQEDFLF

SEQ ID NO: 73

Dryopteris fragraris_DfHAD_wt_nucleic acid sequence

ATGGAGTTCTCTGCCTCTGCTCCTCCTCCTAGGCTAGCCAGTGTCATAATATTGGAG CCTCTCGGCTTCCTCC TCACACCACACTACTCCTCTCAGCTTCCCAAAAAGCTGCTCCGTCGCCTGTTGTGCACTA GAATCTGGCACAG GTATCAGCGAGGCCGCCTTCGCCTGCGTGACGCTGCTATGCTGCTCGCCCAGCTCCCATT CCTAGCTGTGTCT GATCACCCCTGGGCTCTGGACAATCTCGCAAGCCTGCTCCGCCCCACAGCTGTGCGTGCG GTGCCATGGATGC TGCTGCTGCTCGACTTCCTACGAGACGAGCTCCATCTGAAGGTAGTCTGCGCGACCAACT CCTCCCCAGAAGA GCTGCAAGAGCTGCGCCACCAGTTTCCGGCCCTCTTTGCCAAGGTCGATGCCACCGTTTC TTCAGGCGAGGAG GGCGTGGGCAAGCCGTCCGTGCGCTTCCTGCAGGCTGCGTTGGACAAAGCCGGTGTCCAC GCGCAGCAAACCT TGTATCTTGACTCTTTTGACAGCTTGGAGACCATCATGGCTGCACGCTCTCTTGGCATGC ATGCACTATCTGT AGAGCCATGCCACATTGATGAGCTCACCGCCAGGGCCTCTTCCGGCCAGCTAAGAGATGC ACAGCTTATAAGG CGTATTGTGTGCGCCATGCACGGGCCAGCAGTATCTGCAGTTGTGTCGGGCAGTATCACA TCGTCCGGCCCAC AGACAGCAAAGATCGAGGAATTGCCAACAGCTGCTGATAGTCATCTCCGCAGCGCAGCTC TCACTTCTGCTCA GCAGTTTTTCCTCAAAGTTATTGCTCCACATCGTCCTGAGAAGCCATTCGTCCAGCTTCC ATCTCTCACCTCG GAGGGCATCCGAATATACGACACCTTTGCACAGTTTGTCATAGCCGACCTGCTCGACGAC ACCCGCTTCCTAC CCATGCAATCTCCTCCTCCCAATGGGCTCATCACCTTTGTTAACCCAAGCGCGTACCTTG CTGATGATATAAA GAATGGCAACAGCCATATTGTCCCGGGTGTGCAATTTTACGCATCCGATGCGTGCACTCT CATCGACATCCCA CATGACCTAGACACCACCTCCGTTGGCTTGTCAGTACTGCACAAGTTTGGAAAGGTGGAC AAGGACACACTCA ACAAAGTGCTAGACAGAATGCTCGAGCAAGTGAGTGAAGACGACGGCATTCTGCAGGTGT ATTTTGATGTGGA GCGTCCGCGCATCGATCCAGTTGTGGTGGCAAACACGGTGTTTCTGTTCCACTTGGGAAA GAGAGGGCATGAG GTGGCGAGGAGTGAGAAGTTTGTGGAGAGTGTGCTGCTGCAGAGGGCATACGAAGAAGGG ACGTTGTATTACA ACCTGGGGGAAGCATTTTTGGTGAGTGTGGCGAGGCTGGTGCACGAGTTTAAGGAGCACT TTACAAGGAGCGG CATGAGGAGGGCACTGGAGGAGAGGCTAAGAGAGCGGGCAAGGGCGGGCATGCAAGAGAG GGATGATGCGCTG GCGCTAGCCATGCGCATTCGTGCATGCGCTTTGTGTGGCCTGGCCGGAGAGGGCCTCACA AAAGCAGCAGAGC AGGAGCTTTTGCGCCTGCAGTGCAAGTCCAAGGGCTGTTGGGGGTGCCACCCTTTCTATC GCAATGGCAGTAA TGTGCTCAGCTGGATCGGCAGTGAGGCCCTTACCACTGCTTACGCTATTGCTGCGCTACA GCCCATTGATATT TAA SEQ ID NO: 74

Dryopteris fragrans_DfHAD_amino acid sequence

MEFSASAPPPRLASVIILEPLGFLLTPHYSSQLPKKLLRRLLCTRIWHRYQRGRLRL RDAAMLLAQLPFLAVS DHPWALDNLASLLRPTAVRAVPWMLLLLDFLRDELHLKWCATNSSPEELQELRHQFPALF AKVDATVSSGEE GVGKPSVRFLQAALDKAGVHAQQTLYLDSFDSLETIMAARSLGMHALSVEPCHIDELTAR ASSGQLRDAQLIR RIVCAMHGPAVSAW SGSITSSGPQTAKIEELPTAADSHLRSAALTSAQQFFLKVIAPHRPEKPFVQLPSLTS EGIRIYDTFAQFVIADLLDDTRFLPMQSPPPNGLITFVNPSAYLADDIKNGNSHIVPGVQ FYASDACTLIDIP HDLDTTSVGLSVLHKFGKVDKDTLNKVLDRMLEQVSEDDGILQVYFDVERPRIDPVWANT VFLFHLGKRGHE VARSEKFVESVLLQRAYEEGTLYYNLGEAFLVSVARLVHEFKEHFTRSGMRRALEERLRE RARAGMQERDDAL ALAMRIRACALCGLAGEGLTKAAEQELLRLQCKSKGCWGCHPFYRNGSNVLSWIGSEALT TAYAIAALQPIDI DxDxxS

SEQ ID NO: 76

Artificial Sequence_QW motif

QxxxxxW