EP1991689A1 - Verfahren zur herstellung einer feinen chemikalie - Google Patents

Verfahren zur herstellung einer feinen chemikalie

Info

Publication number
EP1991689A1
EP1991689A1 EP07857190A EP07857190A EP1991689A1 EP 1991689 A1 EP1991689 A1 EP 1991689A1 EP 07857190 A EP07857190 A EP 07857190A EP 07857190 A EP07857190 A EP 07857190A EP 1991689 A1 EP1991689 A1 EP 1991689A1
Authority
EP
European Patent Office
Prior art keywords
nucleic acid
acid molecule
polypeptide
column
activity
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP07857190A
Other languages
English (en)
French (fr)
Inventor
Piotr Puzio
Astrid Blau
Tilmann B. Walk
Martijn Gipmans
Volker Haake
Alfons Weig
Gunnar Plesch
Marcus Ebneth
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
BASF Metabolome Solutions GmbH
Original Assignee
Metanomics GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP09156643A priority Critical patent/EP2090662A3/de
Priority claimed from PCT/EP2007/053344 external-priority patent/WO2008034648A1/en
Application filed by Metanomics GmbH filed Critical Metanomics GmbH
Priority to EP07857190A priority patent/EP1991689A1/de
Publication of EP1991689A1 publication Critical patent/EP1991689A1/de
Withdrawn legal-status Critical Current

Links

Definitions

  • the present invention relates to a process for the production of a fine chemical in an organism such as a microorganism, a non-human animal or plant, or in a part thereof.
  • the invention furthermore relates to nucleic acid molecules, antisense, RNAi, snRNA, dsRNA, siRNA, miRNA, ta-siRNA, cosuppression molecule, or ribozyme molecules, or polypeptides, nucleic acid constructs, vectors, antibodies, host cells, plant tissue, propagation material, harvested material, plants, microorganisms as well as agricultural compositions and to their use.
  • Amino acids are used in many branches of industry, including the food, animal feed, cosmetics, pharmaceutical and chemical industries.
  • Amino acids such as D,L-methionine, L-lysine or L-threonine are used in the animal feed industry.
  • the es- sential amino acids valine, leucine, isoleucine, lysine, threonine, methionine, tyrosine, phenylalanine and tryptophane are particularly important for the nutrition of humans and a number of livestock species.
  • Glycine, L-methionine and tryptophane are all used in the pharmaceutical industry.
  • Glutamine, valine, leucine, isoleucine, histidine, argin- ine, proline, serine and alanine are used in the pharmaceutical and cosmetics indus- tries. Threonine, tryptophane and D, L-methionine are widely used feed additives
  • amino acids are suitable for the chemical industry as precursors for the synthesis of synthetic amino acids and proteins, such as N-acetylcysteine, S-carboxymethyl- L-cysteine, (S)-5-hydroxytryptophane and other substances described in Ullmann's Encyclopedia of Industrial Chemistry, vol. A2, pp. 57-97, VCH Weinheim, 1985.
  • strains which are, for example, resistant to antimetabolites, such as, for example, the methionine analogues ⁇ -methylmethionine, ethionine, norleucine, N- acetylnorleucine, S-trifluoromethylhomocysteine, 2-amino-5-heprenoitic acid, selenomethionine, methionine sulfoximine, methoxine, 1-aminocyclopentanecarboxylic acid or which are auxotrophic for metabolites with regulatory importance and which produce sulfur-containing fine chemicals such as, for example, L-methionine.
  • processes developed for the production of methionine have the disadvantage that their yields are too low for being economically exploitable and that they are therefore not
  • Zeh Plant Physiol., Vol. 127, 2001 : 792-802 describes increasing the methionine content in potato plants by inhibiting threonine synthase by what is known as antisense technology. This leads to a reduced threonine synthase activity without the threonine content in the plant being reduced.
  • This technology is highly complex; the enzymatic activity must be inhibited in a very differentiated manner since otherwise auxotrophism for the amino acid occurs and the plant will no longer grow.
  • EP-A-O 271 408 teaches the mutagenesis of plant and selection afterwards with inhibitors of certain enzymes of amino acid biosynthetic pathway.
  • L-methionine is important as methyl group donor for the biosynthesis of, for example, choline, creatine, adrenaline, bases and RNA and DNA, histidine, and for the transmethylation following the formation of S-adenosylmethionine or as a sulfhydryl group donor for the formation of cysteine.
  • L-methionine appears to have a positive effect in depression.
  • the addition of an essential amino acid stimulates protein digestion, which may cause deficiency situations for the second or third limiting amino acid, in particular.
  • feeding experiments for example casein feeding experiments, the additional provision of methionine, which is limiting in casein, has revealed the fatty degeneration of liver, which could only be alleviated after the additional provision of tryptophan.
  • L-methionine is together with lysine or threonine depending on the organism one of the amino acids, which are most frequently limiting.
  • the invention relates to a process for the production of at least one fine chemical selected from the group consisting of: methionine, or, in other words, of the "fine chemical” or "fine chemical of the invention".
  • fine chemical of the invention fine chemical or fine chemical are used herein equally and relate in context of the paragraphs or sections [0014.1.1.1] to [0555.1.1.1] essentially to the metabolite or the metabolites indicated in column 6, Ap- plication No.: 1 of Table I.
  • fine chemical of the invention means methionine in context of the nucleic acid or polypeptide sequences listed in the respective same line of any one of Tables I to IV of Application No.: 1 and indicating in column 6 the metabolite "methionine".
  • methionine or the term "fine chemical” mean in context of the paragraphs or sections [0014.1.1.1] to [0555.1.1.1] at least one chemical com- pound with an activity of the above mentioned methionine, respectively.
  • the terms "fine chemical of the invention”, “fine chemical” or “the fine chemical” mean in context of any of the paragraphs [0014.1.1.1] to [0555.1.1.1] methionine, preferably the L-enantiomer of methionine, in free form or their salts, ester or amides in free form or bound to proteins.
  • fine chemicals as used herein relates to compositions comprising said fine chemical(s), i.e. comprising methionine.
  • methionine as well as the terms “fine chemical'Or “fine chemicals” also encompass the corresponding salts, such as, for example, methionine hydrochloride or methionine sulfate.
  • methionine is intended to encompass the term L-methionine.
  • the present invention relates to a process for the production of methionine, which comprises the following steps:
  • the present invention relates to a process for the pro- duction of methionine, which comprises the following steps:
  • polypeptide comprising a polypeptide, a consensus sequence or at least one polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or of Table IV, respectively; or
  • the methionine synthesized by the organism is recovered or isolated.
  • the process of the invention further comprises reducing, decreasing or deleting the expression or activity of at least one nucleic acid molecule having or encod- ing the activity of at least one nucleic acid molecule represented by the nucleic acid molecule as depicted in Application No.: 1 , column 5 of Table I, and comprising a nucleic acid molecule which is selected from the group consisting of:
  • nucleic acid molecule which, as a result of the degeneracy of the genetic code, can be derived from a polypeptide sequence depicted in Application No.: 1 , column 5 or 7 of Table II;
  • nucleic acid molecule having at least 30 % identity with the nucleic acid molecule sequence of a polynucleotide comprising the nucleic acid molecule shown in Application No.: 1 , column 5 or 7 of Table I;
  • nucleic acid molecule encoding a polypeptide having at least 30 % identity with the amino acid sequence of the polypeptide encoded by the nucleic acid molecule of (a) to (c) and having the activity represented by a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 of Table I;
  • nucleic acid molecule encoding a polypeptide which can be isolated with the aid of monoclonal or polyclonal antibodies made against a polypeptide encoded by one of the nucleic acid molecules of (a) to (e) and having the activity repre- sented by the nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 of Table I;
  • nucleic acid molecule encoding a polypeptide comprising the consensus sequence or one or more polypeptide motifs as shown in Application No.: 1 , column 7 of Table IV and preferably having the activity represented by a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 of Table Il or IV;
  • nucleic acid molecule which comprises a polynucleotide, which is ob- tained by amplifying a cDNA library or a genomic library using the primers in Application No.: 1 , column 7 of Table III which do not start at their 5'-end with the nucleotides ATA and preferably having the activity represented by a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 of Table Il or IV; (j) nucleic acid molecule encoding a polypeptide, the polypeptide being derived by substituting, deleting and/or adding one or more amino acids of the amino acid sequence of the polypeptide encoded by the nucleic acid molecules (a) to (d); and
  • nucleic acid molecule which is obtainable by screening a suitable nu- cleic acid library under stringent hybridization conditions with a probe comprising a complementary sequence of a nucleic acid molecule of (a) or (b) or with a fragment thereof, having at least 15 nt, preferably 20 nt, 30 nt, 50 nt, 100 nt, 200 nt or 500 nt of a nucleic acid molecule complementary to a nucleic acid molecule sequence characterized in (a) to (d) and encoding a polypeptide having the activity represented by a pro- tein comprising a polypeptide as depicted in Application No.: 1 , column 5 of Table II;
  • the process of the invention comprises further reducing, repressing, decreasing or deleting of an expression product of a nucleic acid molecule comprising a nucleic acid molecule as depicted in (a) to (k) above, e.g. a polypeptide comprising a polypeptide as shown in Application No.: 1 , column 5 or 7 of Table Il or of a protein encoded by said nucleic acid molecule.
  • the process of the invention comprises further the reduction of the activity or expression of a polypeptide comprising a polypeptide encoded by the nucleic acid molecule characterized above in an organism or part thereof.
  • the process of the invention comprises further at least one step selected from the group consisting of:
  • nucleic acid molecule encoding a polypeptide having the activity of polypeptide depicted in Application No. 1 , column 5 of Table Il or encoding the expression product of a polynucleotide comprising a nucleic acid molecule as depicted in Application No.: 1 , column 5 or 7 of Table I;
  • RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, cosup- pression molecule, ribozyme, or antisense nucleic acid molecule whereby the RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, cosuppression molecule, ribozyme, or an- tisense nucleic acid molecule comprises a fragment of at least 17 nt with a a homology of at least 50 % to a nucleic acid molecule selected from a group defined in section (a) of this claim.
  • RNAi RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, cosuppression molecule, ribozyme, or antisense nucleic acid molecule characterized in (b) and the ribozyme characterized in (c);
  • nucleic acid molecule comprising a nucleic acid molecule selected from the group defined herein above or defined in section (ab) or (ac) of above or a nucleic acid molecule encoding a polypeptide having at least 50 % identity with the amino acid sequence of the polypeptide encoded by the nucleic acid molecule mentioned above under (a) to (c) and having the activity represented by a protein comprising a polypeptide depicted in Application No. 1 , column 5 of Table Il for inducing a co-suppression of the endogenous expression product;
  • nucleic acid molecule encoding a factor, which binds to a nucleic acid molecule comprising a nucleic acid molecule selected from the group defined herein above or defined in section (ab) or (ac) of this claim conferring the expres- sion of a protein having the activity of a protein encoded by a nucleic acid molecule as characterized herein above;
  • nucleic acid construct capable to recombine with and silence, inactivate, repress or reduces the activity of an endogenous gene comprising a nucleic acid molecule selected from the group defined herein above or defined in section (ab) or (ac) of this claim conferring the expression of a protein encoded by a nu- cleic acid molecule as characterized herein above;
  • a fragment of at least 17 bp of a 3'- or 5'- nucleic acid sequence of a sequences comprising a nucleic acid molecule selected from the group defined herein above or defined in section (ab) or (ac) above with an identity of at least 50 % is used for the reduction of the nucleic acid molecule character- ized above or the polypeptide encoded by said nucleic acid molecule.
  • the reduction or deletion is caused by applying a chemical compound to the non-human-organism.
  • the organism is a transgenic organism selected from the group consisting of a microorganism, a non-human animal and a plant.
  • the plant is selected from the group consisting of Anacardiaceae, Asteraceae, Apiaceae, Betulaceae, Boraginaceae, Brassica- ceae, Bromeliaceae, Caricaceae, Cannabaceae, Convolvulaceae, Chenopodiaceae, Cucurbitaceae, Elaeagnaceae, Ericaceae, Euphorbiaceae, Fabaceae, Geraniaceae, Gramineae, Juglandaceae, Lauraceae, Leguminosae, Linaceae, perennial grass, fod- der crops, vegetables and ornamentals.
  • the microorganism is selected from the group consisting of Actinomycetaceae, Bacillaceae, Brevibacteriaceae, Corynebacteri- aceae, Enterobacteriacae, Gordoniaceae, Micrococcaceae, Mycobacteriaceae, Nocar- diaceae, Pseudomonaceae, Rhizobiaceae, Streptomycetaceae, Chaetomiaceae, Cho- anephoraceae, Cryptococcaceae, Cunninghamellaceae, Demetiaceae, Moniliaceae, Mortierellaceae, Mucoraceae, Pythiaceae, Sacharomycetaceae, Saprolegniaceae, Schizosacharomycetaceae, Sodariaceae, Sporobolomycetaceae, Tuberculariaceae, Adelotheciaceae, Dinophyceae, Ditrichaceae and Prasino
  • the process of the invention further comprises the step, introduction of a RNAi, snRNA, dsRNA, siRNA, miRNA, ta-siRNA, cosuppression molecule, ribozyme, antibody and/or antisense nucleic that has been designed to target the expression product of a gene comprising the nucleic acid molecule as characterized herein above to induce a breakdown of the mRNA of the said gene of interest and thereby silence the gene expression, or of an expression cassette ensuring the expression of the for- mer.
  • the present invention relates to an isolated nucleic acid molecule which comprises a nucleic acid molecule selected from the group consisting of:
  • nucleic acid molecule which comprising a polynucleotide shown in Application No. 1 , column 5 or 7 of Table I B or;
  • nucleic acid molecule comprising a nucleic acid sequence, which, as a result of the degeneracy of the genetic code, can be derived from a polypeptide se- quence depicted in Application No. 1 , column 5 or 7 of Table Il B and having the activity represented by the protein depicted in Application No. 1 , column 5 of Table II;
  • nucleic acid molecule encoding a polypeptide having at least 50 % identity with the amino acid sequence of a polypeptide encoded by the nucleic acid molecule of (a) or (c) and having the activity represented by the protein depicted in Application No. 1 , column 5 of Table Il ;
  • nucleic acid molecule encoding a polypeptide, which is isolated with the aid of monoclonal antibodies against a polypeptide encoded by one of the nucleic acid molecules of (a) to (c) and having the activity represented by the protein depicted in Application No. 1 , column 5 of Table II;
  • nucleic acid molecule which comprises a polynucleotide, which is obtained by amplifying a cDNA library or a genomic library using the primers in Application No.: 1 , column 7 of Table III which do not start at their 5'-end with the nucleotides ATA; and
  • nucleic acid molecule which is obtainable by screening a suitable library under stringent hybridization conditions with a probe comprising one of the sequences of the nucleic acid molecule of (a) to (c) or with a fragment of at least 17 nt of the nucleic acid molecule characterized in any one of (a) to (h) and encoding a polypeptide having the activity represented by the protein depicted in Application No.: 1 , column 5 of Table II; or which comprises a sequence which is complementary thereto;
  • nucleic acid molecule according to (a) to (i) is at least in one or more nucleotides different from the sequence depicted in Application No. 1 , column 5 or 7 of Table I A and preferably which encodes a protein which differs at least in one or more amino acids from the protein sequences depicted in Application No. 1 , column 5 or 7 of Table Il A.
  • the present invention relates to an RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, cosuppression molecule, ribozyme, antibody or an- tisense nucleic acid molecule for the reduction of the activity characterized above or of the activity or expression of a nucleic acid molecule as characterized herein above or a polypeptide encoded by said nucleic acid molecule.
  • the RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, cosuppression molecule, ribozyme, or antisense nucleic acid molecule of the invention comprises a fragment of at least 17 nt of the nucleic acid molecule defined herein above.
  • the present invention relates to a double-stranded RNA (dsRNA), RNAi, snRNA, siRNA, imiRNA, antisense or ta-siRNA molecule or ribozyme, which is able to form a double-stranded ribonucleic acid molecule, whereby a fragment of at least 17 nt of said double-stranded ribonucleic acid molecule has a homology of at least 50 % to a nucleic acid molecule selected from the group of
  • nucleic acid molecule encoding a polypeptide having the activity of polypeptide depicted in Application No. 1 , column 5 or 7 of Table Il or encoding the expression product of a polynucleotide comprising a nucleic acid molecule as depicted in Application No. 1 , column 5 or 7 of Table I.
  • the sense strand and the antisense strand are covalently bound to each other and the antisense strand is essentially the complement of the ,,sense"-RNA strand.
  • the present invention relates to a viral nucleic acid molecule conferring the decline of an RNA molecule conferring the expression of a protein having the activity characterized above or of the activity or expression of a nucleic acid molecule as characterized herein above or a polypeptide encoded by said nucleic acid molecule.
  • the present invention relates to a TILLING primer for the identification of a knock out of a gene comprising a nucleic acid sequence of a nucleic acid molecule as depicted in any one Application No. 1 , column 5 or 7 of Table I.
  • the present invention relates to a dominant-negative mutant of polypeptide comprising a polypeptide as shown in Application No. 1 , column 5 or 7 of Table II.
  • the present invention relates to a nucleic acid molecule encoding the dominant negative mutant defined above.
  • the present invention relates to a nucleic acid con- struct conferring the expression of the RNAi, snRNA, dsRNA, siRNA, miRNA, ta- siRNA, cosuppression molecule, ribozyme, antibody or antisense nucleic acid molecule of the invention, the viral nucleic acid molecule of the invention or the nucleic acid molecule of the invention.
  • the present invention relates to a nucleic acid con- struct comprising the isolated nucleic acid molecule of the invention or the RNAi, snRNA, dsRNA, siRNA, miRNA, ta-siRNA, cosuppression molecule, ribozyme, or antisense nucleic acid molecule of the invention, or the viral nucleic acid molecule of the invention, wherein the nucleic acid molecule is functionally linked to one or more regulatory signals.
  • the present invention relates to a vector comprising the nucleic acid molecule of the invention or the RNAi, snRNA, dsRNA, siRNA, miRNA, ta-siRNA, cosuppression molecule, ribozyme, or antisense nucleic acid molecule of the invention, or the viral nucleic acid molecule of the invention, or the nucleic acid construct of the invention.
  • the nucleic acid molecule is in operable linkage with regulatory sequences for the expression in a prokaryotic or eukaryotic, or in a prokaryotic and eukaryotic host.
  • the present invention relates to a transgenic host cell which has been transformed stably or transiently with the vector of the invention, or the nucleic acid molecule of the invention or the nucleic acid construct of the invention.
  • the host cell is microorganism, a non-human animal or a plant cell.
  • the present invention relates to a host cell or a non- human organism wherein the activity of a protein comprising a polypeptide, a consen- sus sequence or a polypeptide motif as depicted in Application No. 1 , column 5 or 7 of Table II, preferably Table Il B, or IV or a nucleic acid molecule comprising a nucleic acid molecule as depicted in Application No. 1 , column 5 or 7 of Table I, preferably Table I B, is reduced.
  • the present invention relates to a process for producing a polypeptide encoded by a nucleic acid sequence of the invention, the polypeptide being expressed in a host cell of the invention.
  • the host cell is a plant cell selected from the group consisting of Ana- cardiaceae, Asteraceae, Apiaceae, Betulaceae, Boraginaceae, Brassicaceae, Bromeli- aceae, Caricaceae, Cannabaceae, Convolvulaceae, Chenopodiaceae, Cucurbitaceae, Elaeagnaceae, Ericaceae, Euphorbiaceae, Fabaceae, Geraniaceae, Gramineae, Jug- landaceae, Lauraceae, Leguminosae, Linaceae, perennial grass, fodder crops, vegetables and ornamentals or is a microorganism as defined above.
  • the present invention relates to an isolated polypeptide encoded by a nucleic acid molecule of the invention or comprising the polypeptide as depicted in Application No. 1 , column 7 of Table Il B.
  • the present invention relates to an antibody, which specifically binds to the polypeptide of the invention.
  • the present invention relates to a plant tissue, plant, harvested plant material or propagation material of a plant comprising the plant cell of the invention.
  • the present invention relates to a method for screening for an antagonists of the activity as characterized in the process of the invention above or being represented by the polypeptide encoded by the nucleic acid molecule characterized for the process of the invention above:
  • the present invention relates to a process for the identification of a compound conferring increased methionine production in a plant; non- human animal or microorganism, comprising the steps:
  • the present invention relates to a method for the production of an agricultural composition
  • a method for the production of an agricultural composition comprising the steps of the process for the identification of a compound conferring increased methionine production in a plant; non- human animal or microorganism, of the invention and formulating the compound iden- tified said claims in a form acceptable for an application in agriculture.
  • the present invention relates to a composition
  • a composition comprising the protein of the invention, the nucleic acid molecule of the invention, the nucleic acid construct of the invention, the vector of the invention, the antagonist identified according to the method for identification of an antagonist of the invention, the antibody of the invention, the host cell of the invention, the nucleic acid molecule characterized in the process of the invention, the RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, cosuppression molecule, ribozyme, or antisense nucleic acid molecule of the invention and optionally a agricultural acceptable carrier.
  • the present invention relates to a food or feed com- prising the protein of the invention, the nucleic acid molecule of the invention, the nucleic acid construct of the invention, the vector of the invention, the antagonist identified according to the method for identification of an antagonist of the invention, the antibody of the invention, the host cell of the invention, the nucleic acid molecule characterized in the process of the invention, the RNAi, snRNA, dsRNA, siRNA, miRNA, ta-siRNA, cosuppression molecule, ribozyme, or antisense nucleic acid molecule of the invention, the plant, plant tissue, the harvested plant material or propagation material of a plant of the invention.
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps: a) reducing, repressing or deleting of the activity of a oxygen binding protein (CYP86A2) in a non-human organism, and
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • PAKRP1 phragmoplast-associated kinesin-related protein 1
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • CDA1 cytidine deaminase
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps: a) reducing, repressing or deleting of the activity of a beta-galactosidase (BGAL8) in a non-human organism, and
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • the invention relates to a process for the production of methionine, which comprises the following steps:
  • ATPPT1 prenyltransferase
  • the invention relates further to a process for the production of methion- ine, which comprises the following steps:
  • polypeptide comprising a polypeptide, a consensus sequence or at least one polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or of Table IV; or
  • nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Application No.: 1 in column 5 or 7 of Table I B,
  • Table I shows the SEQ ID NOs. of relevant polynucleotides.
  • Table Il shows the SEQ ID NOs. of relevant polypeptides.
  • Table IV shows the SEQ ID NOs. of relevant consensus sequences and relevant polypeptide motifs. In all these tables the abbreviation "A. th.” was used for the organism "Arabidopsis thaliana”.
  • polypeptide as depicted in Table Il or IV also relates to a polypeptide comprising the consensus sequence or at least one polypeptide motif de- picted in Table IV.
  • the molecule which activity is to be reduced according to the process of the invention to provide the increase of methionine e.g. the molecule of I., II., and/or III., above, is in the following the molecule "which activity is to be reduced in the process of the invention".
  • the molecule can for example be a polypeptide or a nucleic acid molecule.
  • the invention relates to a process for the production of the fine chemical as defined above which comprises the following steps:
  • polypeptide comprising a polypeptide selected from the group consisting of SEQ ID NOs 28, 405, 456, 488, 519, 704, 743, 1163,
  • At least one expression product of a nucleic acid molecule comprising a polynucleotide selected from the group consisting of SEQ ID NOs 27, 404, 455, 487, 518, 703, 742, 1 162, 1241 , 1277, 1312, 1417, 1738, 1787, 1891 , 2341 , 2388, 2412, 2560, 3032, 3440, 3858, 117936, 1 17967, 1 17993,
  • IMS3 isopropylmalate synthase
  • ATPPT1 presenilin family protein
  • ribosomal protein ribosomal protein
  • transcription factor WRKY7 transcription factor
  • WRKY7 WRKY7 transcription factor or of a gene comprising a nucleic acid sequence described in column 5 of Table I under Application No.: 1 in Arabidopsis thaliana conferred an increase in the content of methionine in the transformed plants.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 27 in Arabidopsis thaliana conferred an increase in the content of the methionine between 8% and 34% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 404 in Arabidopsis thaliana conferred an increase in the content of the methionine between 8% and 59% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1241 in Arabidopsis thaliana conferred an increase in the content of the methionine between 17% and 53% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1277 in Arabidopsis thaliana conferred an increase in the content of the methionine between 9% and 71 % in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1787 in Arabidopsis thaliana conferred an increase in the content of the methionine between 45% and 91 % in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1891 in Arabidopsis thaliana conferred an increase in the content of the methionine between 16% and 40% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 2388 in Arabidopsis thaliana conferred an increase in the content of the methionine between 28% and 50% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 2412 in Arabidopsis thaliana conferred an increase in the content of the methionine between 19% and 49% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 3858 in Arabidopsis thaliana conferred an increase in the content of the methionine between 9% and 42% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1 17936 in Arabidopsis thaliana conferred an increase in the content of the methionine between 7% and 59% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1 17993 in Arabidopsis thaliana conferred an increase in the content of the methionine between 8% and 53% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1 18042 in Arabidopsis thaliana conferred an increase in the content of the methionine between 19% and 34% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1 18725 in Arabidopsis thaliana conferred an increase in the content of the methionine between 22% and 39% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1 18778 in Arabidopsis thaliana conferred an increase in the content of the methionine between 22% and 63% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1 19300 in Arabidopsis thaliana conferred an increase in the content of the methionine between 112% and 123% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 1 19375 in Arabidopsis thaliana conferred an increase in the content of the methionine between 14% and 43% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 153414 in Arabidopsis thaliana conferred an increase in the content of the methionine between 14% and 92% in a transformed plant as shown in the Examples.
  • the knock out of a gene comprising the nucleic acid sequence SEQ ID NO.: 153470 in Arabidopsis thaliana conferred an increase in the content of the methionine between 15% and 88% in a transformed plant as shown in the Examples.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 27 or polypeptide SEQ ID NO.: 28, respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 404 or polypeptide SEQ ID NO.: 405, respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • nucleic acid molecule SEQ ID NO.: 404 or polypeptide SEQ ID NO.: 405 respectively is reduced or if the activity "oxidoreductase" is reduced in an organism, preferably an increase of methionine between 8% and 59% or more is conferred in said organism.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 455 or polypeptide SEQ ID NO.: 456, respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g.
  • nucleic acid molecule or a polypeptide comprising the nucleic acid or poly- peptide or the consensus sequence or the polypeptide motif depicted in Application No. 1 , Table I, Il or IV, column 7 in the respective same line as the nucleic acid molecule SEQ ID NO.: 455 or polypeptide SEQ ID NO.: 456, respectively is reduced or if the activity "At1g23780-protein" is reduced in an organism, preferably an increase of methionine between 37% and 55% or more is conferred in said organism.
  • the activity of the A thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 487 or polypeptide SEQ ID NO.: 488 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 518 or polypeptide SEQ ID NO.: 519 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 703 or polypeptide SEQ ID NO.: 704, respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 742 or polypeptide SEQ ID NO.: 743 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1162 or polypeptide SEQ ID NO.: 1 163, respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1241 or polypeptide SEQ ID NO.: 1242 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1277 or polypep- tide SEQ ID NO.: 1278 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1312 or polypeptide SEQ ID NO.: 1313 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1417 or polypep- tide SEQ ID NO.: 1418 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1738 or polypeptide SEQ ID NO.: 1739 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1787 or polypeptide SEQ ID NO.: 1788 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1891 or polypep- tide SEQ ID NO.: 1892 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 2341 or polypeptide SEQ ID NO.: 2342 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 2388 or polypeptide SEQ ID NO.: 2389 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 2412 or polypeptide SEQ ID NO.: 2413 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • nucleic acid molecule SEQ ID NO.: 2412 or polypeptide SEQ ID NO.: 2413 is reduced or if the activity "WRKY7 transcription factor" is reduced in an organism, preferably an increase of methionine between 19% and 49% or more is conferred in said organism.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 2560 or polypeptide SEQ ID NO.: 2561 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g.
  • nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif depicted in Application No. 1 , Table I, Il or IV, column 7 in the respective same line as the nucleic acid molecule SEQ ID NO.: 2560 or polypeptide SEQ ID NO.: 2561 , respectively is reduced or if the activity "At4g34770-protein" is reduced in an organism, preferably an increase of methionine between 18% and 61 % or more is conferred in said organism.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 3032 or polypeptide SEQ ID NO.: 3033 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 3440 or polypep- tide SEQ ID NO.: 3441 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 3858 or polypeptide SEQ ID NO.: 3859 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1 17936 or polypep- tide SEQ ID NO.: 1 17937 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 117967 or polypeptide SEQ ID NO.: 1 17968, respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 117993 or polypeptide SEQ ID NO.: 1 17994, respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1 18042 or polypeptide SEQ ID NO.: 1 18043 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • CDA1 cytidine deaminase
  • thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 118180 or polypeptide SEQ ID NO.: 1 18181 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1 18342 or polypeptide SEQ ID NO.: 1 18343 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 118513 or polypeptide SEQ ID NO.: 1 18514 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 118725 or polypep- tide SEQ ID NO.: 1 18726 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 118778 or polypeptide SEQ ID NO.: 1 18779 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 118864 or polypep- tide SEQ ID NO.: 118865 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 1 18893 or polypeptide SEQ ID NO.: 1 18894, respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 118998 or polypeptide SEQ ID NO.: 1 18999 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 119300 or polypeptide SEQ ID NO.: 1 19301 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 119375 or polypeptide SEQ ID NO.: 1 19376, respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 119422 or polypeptide SEQ ID NO.: 119423 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A. thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 153414 or polypeptide SEQ ID NO.: 153415 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • the activity of the A thaliana nucleic acid molecule or a polypeptide comprising the nucleic acid SEQ ID NO.: 153470 or polypeptide SEQ ID NO.: 153471 respectively is reduced or in case in an other organism the activity of the native homolog of said nucleic acid molecule or polypeptide is reduced, e.g. if the activity of a nucleic acid molecule or a polypeptide comprising the nucleic acid or polypeptide or the consensus sequence or the polypeptide motif, depicted in Application No.
  • sequence may relate to polynucleotides, nucleic acids, nucleic acid molecules, peptides, polypeptides and proteins, depending on the context in which the term “sequence” is used.
  • nucleic acid molecule(s) refers to a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides. The terms refer only to the primary structure of the molecule.
  • the terms "gene(s)", “polynucleotide”, “nucleic acid sequence”, “nucleotide sequence”, or “nucleic acid molecule(s)” as used herein include double- and single- stranded DNA and RNA. They also include known types of modifications, for example, methylation, "caps", substitutions of one or more of the naturally occurring nucleotides with an analog.
  • the DNA or RNA sequence comprises a coding sequence encoding the herein defined polypeptide.
  • a "coding sequence” is a nucleotide sequence, which is transcribed into a RNA, e.g.
  • a regulatory RNA such as a miRNA, a ta-siRNA, cosuppression molecule, a RNAi, a ribozyme, etc. or into a mRNA which is translated into a polypeptide when placed under the control of appropriate regulatory sequences.
  • the boundaries of the coding se- quence are determined by a translation start codon at the 5'-terminus and a translation stop codon at the 3'-terminus.
  • a coding sequence can include, but is not limited to mRNA, cDNA, recombinant nucleotide sequences or genomic DNA, while introns may be present as well under certain circumstances.
  • nucleic acid molecule may also encompass the un- translated sequence located at the 3' and at the 5' end of the coding gene region, for example at least 500, preferably 200, especially preferably 100, nucleotides of the sequence upstream of the 5' end of the coding region and at least 100, preferably 50, especially preferably 20, nucleotides of the sequence downstream of the 3' end of the coding gene region.
  • the antisense, RNAi, snRNA, dsRNA, siRNA, miRNA, ta-siRNA, cosuppression molecule, ribozyme etc. technology is used coding regions as well as the 5'- and/or 3'-regions can advantageously be used.
  • Polypeptide refers to a polymer of amino acid (amino acid sequence) and does not refer to a specific length of the molecule. Thus peptides and oligopeptides are included within the definition of polypeptide. This term does also refer to or include post- translational modifications of the polypeptide, for example, glycosylates, acetylations, phosphorylations and the like. Included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatu- ral amino acids, etc.), polypeptides with substituted linkages, as well as other modifications known in the art, both naturally occurring and non-naturally occurring.
  • Table I used in this specification is to be taken to specify the content of Table I A and Table I B.
  • Table II used in this specification is to be taken to specify the content of Table Il A and Table Il B.
  • Table I A used in this speci- fication is to be taken to specify the content of Table I A.
  • Table I B used in this specification is to be taken to specify the content of Table I B.
  • Table Il A used in this specification is to be taken to specify the content of Table Il A.
  • Table Il B used in this specification is to be taken to specify the content of Table Il B.
  • the term “Table I” means Table I B.
  • Table M means Table Il B.
  • the term "organism” as understood herein relates always to a non-human organism, in particular to an animal or plant organism, the whole organism or cell(s) thereof, or to a microorganism. Further, the term “animal” as understood herein relates always to a non-human animal.
  • the overall activity in the volume is reduced, decreased or deleted in cases if the reduction, decrease or deletion is related to the reduction, decrease or deletion of an activity of a gene product, independent whether the amount of gene product or the specific activity of the gene product or both is reduced, decreased or deleted or whether the amount, stability or translation efficacy of the nucleic acid sequence or gene encoding for the gene product is reduced, decreased or deleted.
  • reduction include the change of said property in only parts of the subject of the present invention, for example, the modification can be found in compartment of a cell, like an organelle, or in a part of a plant, like tissue, seed, root, leave, tuber, fruit, flower etc. but is not detectable if the overall subject, i.e. complete cell or plant, is tested.
  • the "reduction”, “repression”, “decrease” or “deletion” is found cellular, thus the term “reduction, decrease or deletion of an activity” or “reduction, decrease or deletion of a metabolite content” relates to the cellular reduction, decrease or deletion compared to the wild type cell.
  • the terms “reduction”, “repression”, “decrease” or “deletion” include the change of said property only during different growth phases of the organism used in the inventive process, for example the reduction, repression, decrease or deletion takes place only during the seed growth or during blooming.
  • the terms include a transitional reduction, decrease or deletion for example because the used method, e.g. the an- tisense, RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, cosuppression molecule, or ribozyme, is not stable integrated in the genome of the organism or the reduction, decrease, repression or deletion is under control of a regulatory or inducible element, e.g. a chemical or otherwise inducible promoter, and has therefore only a transient effect.
  • a regulatory or inducible element e.g. a chemical or otherwise inducible promoter
  • the term “reduction”, “repression”, “decrease” or “deletion” means that the specific activity of a gene product, an enzyme or other protein or a regulatory RNA as well as the amount of a compound or metabolite, e.g. of a polypeptide, a nucleic acid molecule, a fine chemical or an encoding imRNA or DNA, can be reduced, decreased or deleted in a specific volume.
  • the terms “reduction”, “repression”, “decrease” or “deletion” include that the reason for said “reduction”, “repression”, “decrease” or “deletion” can be a chemical compound that is administered to the organism or part thereof.
  • Reduction is also understood as meaning the modification of the substrate specificity as can be expressed for example, by the kcat/Km value.
  • the function or activity e.g. the enzymatic activity or the "biological activity”
  • the function or activity is reduced by at least 10%, advantageously 20%, preferably 30%, especially preferably 40%, 50% or 60%, very especially preferably 70%, 80%, 85% or 90% or more, very especially preferably are 95%, more preferably are 99% or more in comparison to the control, reference or wild type.
  • Most preferably the reduction, decrease or deletion in activity amounts to essentially 100%.
  • a particularly advantageous embodiment is the inactivation of the function of a compound, e.g. a polypeptide or a nucleic acid molecule.
  • the reduction, repression or deletion of the expression level or of the activity leads to an increase of the fine chemical content of 10%, 20%, 30%, 40%, 50%, 100%, 150% or 200% or more, preferably of 250% or 300% or more, particularly preferably of 350% or 400% or more, most particularly preferably of 500% or 600% w/w, or more, in an specific volume, e.g. in a cell, a tissue, an organ, an organism, or a part thereof in com- parison to the reference or wild type.
  • activity of a compound refers to the function of a compound in a biological system such as a cell, an organ or an organism.
  • activity of a compound refers to the enzymatic function, regulatory function or its function as binding partner, transporter, regulator, or carrier, etc of a compound.
  • biological activity refers to an activity selected from the group consisting of:
  • IMS3 2-isopropylmalate synthase
  • 3-isopropylmalate dehydrogenase / oxidore- ductase anion exchanger, aspartyl protease, AT1 G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP-dependent peptidase/ ATPase/ nucleoside- triphosphatase/ serine- type endopeptidase, ATR2-protein, beta-galactosidase (BGAL8), calmodulin binding protein / translation elongation factor, choline kinase (AT
  • the terms “enhance”, “increase”, “decrease”, “repress” or “reduce” or similar terms include the change or the modulation of said property in only one or some parts as well as in all parts of the subject of the present invention.
  • the modification can be found in compartment of a cell, like an organelle, or in a part of a plant, like a tissue, seed, root, leave, fruit, tuber, flower etc. but is not detectable if the overall subject, i.e. complete cell or plant, is tested.
  • the change or the modulation of said property is found at least cellular, thus the term "decrease of an activity” or "increase of a metabolite content” relates to at least a cellular increase compared to the wild type cell.
  • More preferred is the finding that a change or a modulation of said property is found in more than one part of an organism, particularly of a plant.
  • the change or the modulation of said property is found in a tissue, seed, root, fruit, tuber, leave and/or flower of a plant produced according to the process of the present invention.
  • this process further comprises the step of recovering the fine chemical, which is synthesized by the organism from the organism and/or from the medium used for the growth or maintenance of the organism.
  • the term "recovering” means the isolation of the fine chemical in different purities, that means on the one hand harvesting of the biological material, which contains the fine chemical without further purification and on the other hand purities of the fine chemical between 5% and 100% w/w purity, preferred purities are in the range of 10% and 95% w/w, e.g. between 20%, 30%, 40% or 50% and 60%, 70%, 80% or 90% w/w. In one embodiment, the purity of the fine chemical is 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 99% w/w.
  • the process for the production of the fine chemical leads to an enhanced production of the fine chemical in comparison to the appropiate control organ- ism or tissue.
  • the terms “enhanced” or “increase” mean a 10%, 20%, 30%, 40% or 50% or higher, preferably at least a 60%, 70%, 80%, 90% or 100% or higher, more preferably 150%, 200%, 300%, 400% or 500% or higher production of the fine chemical in an specific volume, e.g. in a cell, a tissue, an organ, an organism, or a part thereof in comparison to the reference or wild type.
  • the increase is calculated as in the examples shown.
  • the term increase means the increase in amount in relation to the weight of the organism or part thereof (w/w).
  • the fine chemical increased and recovered in the process of the present invention is methionine.
  • the increase of said fine chemical is preferably found in whole plants or in leaves, flowers, seeds, fruits, roots, and/or tubers of a plant produced according to the process of the present invention. This does not exclude that the actual increase of the fine chemical itself can occur in an organelles, like plastids, mitochondria, peroxisomen, glyoxysomes or others.
  • the term "reference”, "control” or “wild type” mean an organism without the aforementioned modification of the expression or activity of an expression product of a nucleic acid molecule comprising a polynucleotide indicated in Table I, Application No.: 1 , column 5 or 7 or of the activity of a protein having the activity of a polypeptide comprising a polypeptide indicated in Table Il or IV, Application No.: 1 , column 5 or 7, or of the activity of a protein encoded by nucleic acid molecule comprising a nucleic acid molecule indicated in Table I, Application No.: 1 , column 5 or 7.
  • a "reference”, “control” or “wild type” is in particular a cell, a tissue, an organ, an organism, or a part thereof which was not produced according to the process of the invention.
  • wild type can be a cell or a part of organisms such as an organelle or tissue, or an organism, in particular a microorganism or a plant, which was not modified or treated according to the herein described process according to the invention.
  • the cell or a part of organisms such as an organelle or a tissue, or an organism, in particular a microorgan- ism or a plant used as wild type
  • control or reference corresponds to the cell, organism or part thereof as much as possible and is in any other property but in the result of the process of the invention as identical to the subject matter of the invention as possible.
  • the wild type, control or reference is treated identically or as identical as possible, saying that only conditions or properties might be different which do not influence the quality of the tested property.
  • analogous conditions means that all conditions such as, for example, culture or growing conditions, assay conditions (such as buffer composition, temperature, substrates, pathogen strain, concentrations and the like) are kept identical between the experiments to be compared.
  • the "reference”, "control”, or “wild type” is preferably a subject, e.g. an organelle, a cell, a tissue, an organism, in particular a plant or a microorganism, which was not modified or treated according to the herein described process of the invention and is in any other property as similar to the subject matter of the invention as possible.
  • the reference, control or wild type is in its genome, transcriptome, proteome or metabolome as similar as possible to the subject of the present invention.
  • the term "reference-" "control-” or “wild type-”-organelle, -cell, -tissue or -organism, in particular plant or microorganism relates to an organelle, cell, tissue or organism, in particular plant or micro-organism, which is nearly genetically identical to the organelle, cell, tissue or organism, in particular microorganism or plant, of the present invention or a part thereof preferably 95%, more preferred are 98%, even more preferred are 99,00%, in particular 99,10%, 99,30%, 99,50%, 99,70%, 99,90%, 99,99%, 99, 999% or more.
  • the "reference”, “control”, or “wild type” is preferably a subject, e.g. an organelle, a cell, a tissue, an organism, which is genetically identical to the organism, cell organelle used according to the process of the invention except that nucleic acid molecules or the gene product encoded by them are changed or modified according to the inventive process.
  • a control, reference or wild type differing from the subject of the present invention only by not being subject of the process of the invention can not be provided
  • a control, reference or wild type can be an organism in which the cause for the modulation of the activity conferring the increase of the fine chemical as described herein has been switched back or off, e.g. by complementation of responsible reduced gene product, e.g. by stable or transient (over)expression, by activation of an activator or agonist, by inactivation of an inhibitor or antagonist, by adding active compounds as e.g. hormones, by introducing enhancers etc.
  • the reference and the subject matter of the invention are compared after standardization and normalization, e.g. to the amount of total RNA, DNA, or protein or activity or expression of reference genes, like housekeeping genes, such as certain actin or ubiquitin genes.
  • the reference, control or wild type differs form the subject of the present invention only in the cellular activity of the polypeptide or RNA used in the process of the invention, e.g. as result of a reduction, decrease or deletion in the level of the nucleic acid molecule of the present invention or a reduction, decrease or deletion of the specific activity of the polypeptide or RNA used in the process of the invention, e.g. by the expression level or activity of protein or RNA, that means by reduction or inhibition of its biological activity and/or of its biochemical or genetical causes.
  • expression refers to the transcription and/or translation of a codogenic gene segment or gene.
  • the resulting product is a imRNA or a protein.
  • expression products can also include functional RNAs such as, for example, antisense, tRNAs, snRNAs, rRNAs, dsRNAs, siRNAs, imiRNAs, ta-siRNA, co- suppression molecules, ribozymes etc.
  • Expression may be systemic, local or temporal, for example limited to certain cell types, tissues organs or time periods.
  • RNA e.g. rRNA, tRNA, imiRNA, dsRNA, snRNA, ta-siRNA, siRNA
  • RNA messenger RNA
  • expression means the expression of a gene with or without the subsequent translation of the latter into a protein.
  • expression on RNA level can be detected by methods well known, e.g. Northern blotting, array hybridizations, qRT
  • polypeptide level can be detected by methods well known, e.g. Western blotting or other immuno assays.
  • nucleic acid molecule as depicted in Application No.: 1 , column 5 or 7 of Table I is a polynucleotide which confers essentially the activity of a nucleic acid molecule as depicted in Application No.: 1 , column 5 of Table I.
  • a protein or polypeptide has the activity of a polypeptide as depicted in Application No.: 1 , column 5 of Table Il if the reduction, repression, decrease or deletion of its activity mediates the increase of methionine.
  • a protein or polypeptide has the activity of a polypeptide as depicted in Application No.: 1 , column 5 of Table Il if the reduction, repression, decrease or deletion of its activity mediates the increase of methionine as indicated in the respective same line of Table Il or IV in column 6.
  • a nucleic acid molecule or polynucleotide has the activity of a nucleic acid molecule as depicted in Application No.: 1 , column 5 of Table I" if the reduction, repression, decrease or deletion of its expression mediates the increase of methionine.
  • a nucleic acid molecule or polynucleotide has the activity of a nucleic acid molecule as depicted in Application No.: 1 , column 5 or 7 of Table I" if the reduction, repression, decrease or deletion of its expression mediates the increase of methionine as indicated in the respective same line of Table I in column 6.
  • the reduction, repression or deletion of the activity of such an aforementioned protein or polypeptide or of the expression product of such an aforementioned nucleic acid molecule or sequence means a reduction of the expres- sion level or activity of the gene product or the polypeptide, for example the enzymatic or biological activity of the polypeptide, of at least 10% preferably 20%, 30%, 40% or 50%, particularly preferably 60% 70% or 80%, most particularly preferably 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% in comparison to the original endogenous expression level of the expression product or to the original endogenous activity of an expression product or polypeptide comprising or being encoded by a nucleic acid molecule as indicated in Application No.: 1 , column 5 or 7 of Table I or comprising a polypeptide as indicated in Application No.: 1 , column 5 or 7 of Table Il or IV or the endogenous homologue or equivalent thereof.
  • nucleic acid molecule has "activity of a nucleic acid molecule as depicted in Application No.: 1 , column 5 of Table I" in a complementation assay.
  • a gene is a functional homologue of a nucleic acid molecule depicted in columns 5 or 7, in particular depicted in column 5, a complementation assay in a microorganism or a plant can be performed.
  • a plant lacking the activity of the gene e.g. a Arabidopsis thaliana strain in which a nucleic acid molecule comprising the nucleic acid molecule has been knocked out, in particular deleted or interrupted, can be transformed with the respective nucleic acid molecule in question, e.g. a gene or homologue, under control of a suitable promoter, e.g. in a suitable vector.
  • the promoter may either confer constitutive or transient or tissue or development specific or inducible expression.
  • the promotor may be similar or identical in spatial and temporal activity to the promoter of the gene, which has been knock out, deleted or interrupted.
  • the nucleic acid molecule in question e.g. the gene or the homologue to be tested preferably comprises the complete coding region either with or without introns(s).
  • Transformed plants are analyzed for the presence of the respective construct and the expression of the nucleic acid molecule in question, e.g. the gene or homologue, or its expression product. Plants exhibiting expression of the gene or homologue are compared to wild type plants.
  • the transgenic plant, comprising a knockout mutation and expressing the respective gene or homologue is essentially identical to wild type controls with regard to the change in the concentration of the fine chemical indicated in Table I, column 6 in the same line as the knocked out molecule is shown.
  • a qualified complementation assay is for example described in lba K (1993) Journal of Biological Chemistry 268 (32) pp24099-24105, Bon Rush G et al (2003) Plant Growth. Plant Cell 15 pp 1020-1033, or in Gachotte D et al (1995) Plant Journal 8 (3) pp 407-416.
  • At1g07890 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as L-ascorbate peroxidase (APX1 ).
  • the process of the present invention comprises the reduction of a gene product with the activity of a "L-ascorbate peroxidase (APX1 )" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "L-ascorbate peroxidase (APX1 )", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • APX1 L-ascorbate peroxidase
  • the sequence of At1g22950 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as oxidoreductase.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "oxidoreductase” from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "oxidoreduc- tase", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the process of the present invention comprises the reduction of a gene product with the activity of a "At1g23780-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "At1g23780- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the process of the present invention comprises the reduction of a gene product with the activity of a "At1g27695-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "At1g27695- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the process of the present invention comprises the reduction of a gene product with the activity of a "kinase” from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "kinase", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At1g80920 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as heat shock protein binding protein / unfolded protein binding protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "heat shock protein binding protein / unfolded protein binding protein” from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "heat shock protein binding protein / unfolded protein binding protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the process of the present invention comprises the reduction of a gene product with the activity of a "ATP-dependent peptidase/ ATPase/ nucleoside- triphosphatase/ serine-type endopeptidase" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "ATP- dependent peptidase/ ATPase/ nucleoside- triphosphatase/ serine-type endopeptidase", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At3g06450 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "anion exchanger" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "anion exchanger", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the process of the present invention comprises the reduction of a gene product with the activity of a "At3g12850-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "At3g 12850- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as At3g20380-protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "At3g20380-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "At3g20380- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At3g24090 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as glutamine-fructose-6-phosphate transaminase.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "glutamine-fructose-6-phosphate transaminase" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "glutamine- fructose-6-phosphate transaminase", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At3g55990 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102-5), and its activity is described as At3g55990-protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "At3g55990-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "At3g55990- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At3g59340 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as At3g59340-protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "At3g59340-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • At3g59340 for the production of the methionine in particular for increasing the amount of the L-enantiomer of said amino acids in free or bound form in an organism or a part thereof, as mentioned.
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "At3g59340- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At4g00360 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as oxygen binding protein (CYP86A2) .
  • the process of the present invention comprises the reduction of a gene product with the activity of a “oxygen binding protein (CYP86A2) " from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "oxygen binding protein (CYP86A2) ", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At4g10350 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102-5), and its activity is described as At4g10350-protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "At4g10350-protein” from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "At4g10350- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At4g14150 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as phragmoplast-associated kinesin-related protein 1 (PAKRP1 ).
  • the process of the present invention comprises the reduction of a gene product with the activity of a "phragmoplast-associated kinesin- related protein 1 (PAKRP1 )" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "phragmo- plast-associated kinesin-related protein 1 (PAKRP1 )", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • PAKRP1 plasmin-associated kinesin-related protein 1
  • the sequence of At4g16141 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102-5), and its activity is described as At4g 16141 -protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "At4g 16141 -protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "At4g16141- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At4g24240 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as WRKY7 transcription factor.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "WRKY7 transcription factor" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "WRKY7 transcription factor", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At4g34770 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as At4g34770-protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "At4g34770-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of (a) a gene product of a gene comprising the nucleic acid molecule as shown in Application No. 1 , column 5 of Table I and being depicted in the same respective line as said At4g34770 or a functional equivalent or a homologue thereof as shown in Application No.: 1 , column 7 of Table I, preferably a homologue or func- tional equivalent as shown in Application No.: 1 , column 7 of Table I B, and being depicted in the same respective line as said At4g34770; or
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "At4g34770- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At5g23010 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as 2-isopropylmalate synthase (IMS3).
  • IMS3 2-isopropylmalate synthase
  • the process of the present invention comprises the reduction of a gene product with the activity of a "2-isopropylmalate synthase (IMS3)" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • IMS3 2-isopropylmalate synthase
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "2- isopropylmalate synthase (IMS3)", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • IMS3 2- isopropylmalate synthase
  • the process of the present invention comprises the reduction of a gene product with the activity of a "aspartyl protease” from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "aspartyl protease", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At5g60390 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as calmodulin binding protein / translation elongation factor.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "calmodulin binding protein / transla- tion elongation factor" from Arabidopsis thaliana or its functional equivalent or its ho- molog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "calmodulin binding protein / translation elongation factor", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At1g08700 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102-5), and its activity is described as presenilin family protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "presenilin family protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "presenilin family protein", preferably it is the molecule of section (a) or (b) of this paragraph
  • AT1G13880 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as AT1G13880-protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "AT1G13880-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "AT1G13880- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • AT1G13880- protein preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of AT1 G71697 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as choline kinase (ATCK1 ).
  • the process of the present invention comprises the reduction of a gene product with the activity of a "choline kinase (ATCK1 )" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "choline kinase (ATCK1 )", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At2g19570 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as cytidine deaminase (CDA1 ).
  • the process of the present invention comprises the reduction of a gene product with the activity of a "cytidine deaminase (CDA1 )" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "cytidine deaminase (CDA1 )", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • CDA1 cytidine deaminase
  • the process of the present invention comprises the reduction of a gene product with the activity of a "ATP binding protein (CPN60A)" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "ATP binding protein (CPN60A)", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • ATP binding protein CPN60A
  • BGAL8 beta-galactosidase
  • the process of the present invention comprises the reduction of a gene product with the activity of a "beta-galactosidase (BGAL8)" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "beta- galactosidase (BGAL8)", preferably it is the molecule of section (a) or (b) of this para- graph [0024.1.1.1].
  • BGAL8 beta- galactosidase
  • PMR6 pectate lyase protein / powdery mildew suscep- tibility protein
  • the process of the present invention comprises the reduction of a gene product with the activity of a "pectate lyase protein / powdery mildew susceptibility protein (PMR6)" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "pectate lyase protein / powdery mildew susceptibility protein (PMR6)", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of AT3G55750 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102-5), and its activity is described as ribosomal protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "ribosomal protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "ribosomal protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • ribosomal protein preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of AT3G60770 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as ribosomal protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "ribosomal protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "ribosomal protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • ribosomal protein preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At4g14713 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as AT4G14713-protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "AT4G14713-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "AT4G14713- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of AT4G30210 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as ATR2-protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "ATR2-protein” from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "ATR2- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • ATR2- protein preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At5g02330 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as DC1 domain-containing protein / protein-binding protein / zinc ion binding protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "DC1 domain-containing protein / pro- tein-binding protein / zinc ion binding protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "DC1 domain- containing protein / protein-binding protein / zinc ion binding protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of AT5G14200 from Arabidopsis thaliana e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102-5), and its activity is described as 3- isopropylmalate dehydrogenase / oxidoreductase.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "3-isopropylmalate dehydrogenase / oxidoreductase" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "3- isopropylmalate dehydrogenase / oxidoreductase", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the process of the present invention comprises the reduction of a gene product with the activity of a "AT5G26850-protein" from Arabidop- s/s thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "AT5G26850- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the process of the present invention comprises the reduction of a gene product with the activity of a "transcription factor" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of (a) a gene product of a gene comprising the nucleic acid molecule as shown in Application No. 1 , column 5 of Table I and being depicted in the same respective line as said AT5G39860 or a functional equivalent or a homologue thereof as shown in Application No.: 1 , column 7 of Table I, preferably a homologue or func- tional equivalent as shown in Application No.: 1 , column 7 of Table I B, and being depicted in the same respective line as said AT5G39860; or
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "transcription factor", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • transcription factor preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of AT4G20940 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as AT4G20940-protein.
  • the process of the present invention comprises the reduction of a gene product with the activity of a "AT4G20940-protein" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "AT4G20940- protein", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • the sequence of At4g23660 from Arabidopsis thaliana, e.g. as shown in application No.: 1 , column 5 of Table I, has been published in the TAIR database http://www.arabidopsis.org (Huala, E. et al., Nucleic Acids Res. 2001 Vol. 29(1 ), 102- 5), and its activity is described as prenyltransferase (ATPPT1 ).
  • the process of the present invention comprises the reduction of a gene product with the activity of a "prenyltransferase (ATPPT1 )" from Arabidopsis thaliana or its functional equivalent or its homolog, e.g. the reduction of
  • the molecule which activity is to be reduced in the process of the invention is the gene product with an activity described as "prenyltransferase (ATPPT1 )", preferably it is the molecule of section (a) or (b) of this paragraph [0024.1.1.1].
  • homologues of the present gene products in particular homologues of a gene product which is encoded by or which is comprising a nucleic acid molecule as shown in Application No.: 1 , column 7 of Table I, or a polypeptide comprising the polypeptide , a consensus sequence or a polypeptide motif as shown in Application No.: 1 , column 7 of Table Il or IV, can be derived from any organisms as long as the homologue confers the herein mentioned activity, i.e. it is a functional equivalent of said molecules. In particular, the homologue confers an increase in the fine chemical amount or content after its reduction or deletion.
  • the term "homologue” relates to the se- quence of an organism having preferably the highest or essentially the highest sequence homology to the herein mentioned or listed sequences of all expressed sequences of said organism.
  • the person skilled in the art knows how to find, identify and confirm, that, preferably, a putative homologue has said the-fine-chemical-increasing activity, e.g. as described herein.
  • the biological function or activity in an organism essentially relates or corresponds to the activity or function as described for the genes mentioned in paragraph [0024.1.1.1], for example to at least one of the protein(s) indicated in Application No.: 1 of Table II, Column 5.
  • the homologue or the functional equivalent comprises the sequence of a polypeptide encoded by a nucleic acid molecule comprising a sequence indicated in Application No.: 1 of Table I, Column 7 or a polypeptide sequence, a consensus sequence or a polypeptide motif indicated in Application No.: 1 of Table Il or IV, Column 7 or it is the expression product of a nucleic acid molecule comprising a polynucleotide indicated in Application No.: 1 of Table I, Column 7,.
  • the activity of a protein or polypeptide or a nucleic acid molecule or sequence encoding such protein or polypeptide e.g. an activity selected from the group consisting of 2-isopropylmalate synthase (IMS3), 3- isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1 G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP-dependent peptidase/ AT- Pase/ nucleoside- triphosphatase/ serine-type endo
  • IMS3 2-is
  • the homolog of any one of the polypeptides indicated in Table II, application No.: 1 , column 5 is derived from an Eukaryot and has a sequence identity of at least 50% and preferably has essentially the same or a similar activity as described in [0024.1.1.1], however its reduction of expression or activity confers an increase in the content of methionine, respectively, in the organisms or a part thereof.
  • the homolog of any one of the polypeptides indicated in Table II, application No.: 1 , column 5 is derived from a plant, preferably from a plant selected from the group consisting of Nacardiaceae, Asteraceae, Apiaceae, Betulaceae, Bo- raginaceae, Brassicaceae, Bromeliaceae, Caricaceae, Cannabaceae, Convolvulaceae, Chenopodiaceae, Cucurbitaceae, Elaeagnaceae, Ericaceae, Euphorbiaceae, Fa- baceae, Geraniaceae, Gramineae, Juglandaceae, Lauraceae, Leguminosae, Linaceae, perennial grass, fodder crops, vegetables and ornamentals and has a sequence identiy of at least 50% and preferably has essentially the same or a essentially similar activity as described in [0024.1.1.1], however at least its reduction of expression or activity confers an increase in the content of
  • the homolog of any one of the polypeptides indicated in Table II, application No.: 1 , column 5 is derived from a crop plant and has a sequence identiy of at least 30% and preferably has essentially the same or a similar activity as described in [0024.1.1.1], however at least an reduction of expression or activity confers an increase in the content of methionine, repectively, in the organisms, or a part thereof.
  • the homolog of any one of the polypeptides indicated in Table II, application No.: 1 , column 5 is derived from a microorganism, preferably from a micro- organism selected from the group consisiting of Actinomycetaceae, Bacillaceae, Brevi- bacteriaceae, Corynebacteriaceae, Enterobacteriacae, Gordoniaceae, Micrococca- ceae, Mycobacteriaceae, Nocardiaceae, Pseudomonaceae, Rhizobiaceae, Strepto- mycetaceae, Chaetomiaceae, Choanephoraceae, Cryptococcaceae, Cunning- hamellaceae, Demetiaceae, Moniliaceae, Mortierellaceae, Mucoraceae, Pythiaceae, Sacharomycetaceae, Saprolegniaceae, Schizosacharomycetaceae, Sodariaceae, Sporobolomycetaceae
  • the molecule which activity is to be reduced in the process of the invention is the molecule of (a) or (b) of paragraph [0024.1.1.1], [0025.1.1.1] or of paragraph [0027.1.1.1].
  • a homolog or a functional equivalent of a polypeptide as indicated in Table II, column 3 or column 5 may be a polypeptide encoded by a nucleic acid molecule comprising a polynucleotide as indicated in Table I, column 7 in the same line, or may be a polypeptide comprising a polypeptide indicated in Table II, column 7, or one or more polypeptide motifs indicated in Table IV, column 7, or the consensus sequence as indicated in Table IV, column 7 in the same line as the polypeptide indi- cated in Table II, column 3 or column 5.
  • column 5 may be a nucleic acid molecule comprising a polynucleotide indicated in Table I, column 7 in the same line, or nucleic acid molecule encoding a polypeptide comprising a polypeptide indicated in Table II, column 7, or the consensus se- quence or polypeptide motifs indicated in Table IV, column 7 in the same line as the nucleic acid molecule indicated in Table I, column 3 or column 5.
  • a decreased or reduced activity of the molecule which activity is to be reduced in the process of the invention manifests itself in an increase in the respective fine chemical.
  • the fine chemical's amount in a cell preferably in a tissue, more preferred in an organism as a plant or a microorganism or part thereof, is increased by 3% or more, especially preferably are 10% or more, very especially pref- erably are more than 30% and most preferably are 70% or more, such as 100%, 300% or 500% or more.
  • the respective fine chemical can be contained in the organism either in its free form and/or bound to proteins or polypeptides or mixtures thereof or can advantageously be extracted.
  • compositions or mixtures of various fine chemicals can be produced, e.g. comprising one or more further distinct amino acids, in particular rare or healthy amino acids, fatty acids, vitamins, hormones, sugars, lipids, etc.
  • the process comprises reducing or deleting the expression or activity of at least one nucleic acid molecule having or encoding the activity of at least one nucleic acid molecule represented by the nucleic acid molecule as depicted in Application No.: 1 , column 5 of Table I, and wherein the nucleic acid molecule comprises a nucleic acid molecule selected from the group consisting of:
  • nucleic acid molecule encoding the polypeptide shown in Application No.: 1 , column 5 or 7 of Table Il or IV;
  • nucleic acid sequence which, as a result of the degeneracy of the genetic code, can be derived from a polypeptide sequence depicted in Application No.:
  • nucleic acid molecule having at least 30%, 40%, 50%, 60%, 70 %, 80%, 90%, 95%, 97%, 98%, 99%, 99,9% identity with the nucleic acid molecule sequence of a polynucleotide comprising the nucleic acid molecule shown in Ap- plication No.: 1 , column 5 or 7 of Table I; e) a nucleic acid molecule encoding a polypeptide having at least 30%, 40%, 50%, 60%, 70 %, 80%, 90%, 95%, 97%, 98%, 99%, 99,9% identity with the amino acid sequence of the polypeptide encoded by the nucleic acid molecule of (a) to (c) and having the activity represented by a protein as depicted in Ap- plication No.: 1 , column 5 of Table II;
  • nucleic acid molecule encoding a polypeptide which is isolated with the aid of monoclonal or polyclonal antibodies made against a polypeptide encoded by one of the nucleic acid molecules of (a) to (e) and having the activity represented by the protein as depicted in Application No.: 1 , column 5 of Table Il ;
  • nucleic acid molecule encoding a polypeptide comprising the consensus sequence or one or more polypeptide motifs shown in the corresponding lane of Application No.: 1 , column 7 of Table IV and preferably having the activity represented by a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 of Table I;
  • nucleic acid molecule which comprises a polynucleotide, which is obtained by amplifying a cDNA library or a genomic library using primers in Application No.: 1 , column 7 of Table III which do not start at their 5 prime end with the nucleotides ATA; and preferably having the activity represented by a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 of Table I;
  • nucleic acid molecule encoding a polypeptide having the activity represented by the protein as depicted in Application No.: 1 , column 5 of Table Il ;
  • nucleic acid molecule which is obtainable by screening a suitable nucleic acid library under stringent hybridization conditions with a probe comprising a complementary sequence of a nucleic acid molecule of (a) or (b) or with a fragment thereof having at least 15 nt of a nucleic acid molecule complementary to a nucleic acid molecule sequence characterized in (a) to (d) and encoding a polypeptide having the activity represented by a protein as depicted in Application No.: 1 , column 5 of Table Il ;
  • nucleic acid molecule or of a protein encoded by said nucleic acid molecule.
  • the term "molecule which activity is to be reduced in the process of the invention” refers to above said nucleic acid molecule comprising at least one of said nucleic acid molecules a) to j) according to this paragraph [0033.1.1.1].
  • said nucleic acid molecule or said polypeptide depicted in Application No.: 1 , column 5 or 7 of Table I, Il or IV is a novel nucleic acid molecule or a novel polypeptide as depicted in Application No.: 1 , column 5 or 7 of Table I B or Il B.
  • a polypeptide or a nucleic acid molecule in particular a nucleic acid molecule comprising the nucleic acid molecule described in Application No. 1 , column 5 or 7 of Table I or a polypeptide comprising a polypeptide as described in Application No. 1 , column 5 or 7 of Table Il or IV, or a functional homolog of said nucleic acid molecule or polypeptide, can be manipulated to directly or indirectly affect the yield, production and/or production efficiency of a amino acids, in particular of methionine.
  • the molecule number or the specific activity of the polypeptide which activity is to be reduced in the process of the invention or processed by polypeptide which activity is to be reduced in the process of the invention or the molecule number proc- essed by or expressed by the nucleic acid molecule which activity is to be reduced in the process of the invention may be reduced, decreased or deleted.
  • reduction of a biological function refers, for example, to the quantitative re-duction in a binding capacity or binding strength of a protein to a substrate in an organism, a tissue, a cell or a cell compartment in comparison with the wild type of the same genus and species to which this method has not been applied, under otherwise identical conditions (such as, for example, culture conditions, age of the plants and the like).
  • Binding partners for the protein can be identified in the manner with which the skilled worker is familiar, for example by the yeast 2-hybrid system.
  • the reduction, repression, decrease, deletion or modulation according to this invention can be conferred by the (e.g. transgenic) expression of a antisense nucleic acid molecule, an RNAi, a snRNA, a dsRNA, a siRNA, a imiRNA, a ta-siRNA, a cosuppression molecule, a ribozyme or of an antibody, an inhibitor or of an other molecule inhibiting the expression or activity of the expression product of the nucleic acid molecule which activity is to be reduced, decreased or deleted in the process of the invention.
  • the reduction, repression, decrease, deletion or modulation according to this invention can be conferred by the (e.g.
  • nucleic acid molecule comprising a polynucleotide encoding antisense nucleic acid molecule, RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, a cosuppression molecule, ribozyme or of an antibody against the nucleic acid molecule or the polypeptide which activity is to be reduced in the process of the invention.
  • the reduction, repression, decrease, deletion or modulation according to this invention can be to a stable mutation in the corresponding endogenous gene encoding the nucleic acid molecule to be reduced, decreased or deleted in the process of the invention, e.g. of a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7 of Table I.
  • the reduction, repression, decrease, deletion or modulation according to this invention can be a modulation of the expression or of the behaviour of a gene conferring the expression of the polypeptide to be reduced, decreased, repressed or deleted according to the process of the invention, e.g. of a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV
  • Said expression may be constitutive, e.g. due to a stable, permanent, systemic, local or temporal expression, for example limited to certain cell types, tissues organs or time periods.
  • the reduction, repression, decrease, deletion or modulation according to this invention can be transient, e.g. due to an transient transformation, a transiently active promotor or temporary addition of a modulator, such as an antagonist, inhibitor or inductor, e.g. after transformation with an inducible construct carrying the double- stranded RNA nucleic acid molecule (dsRNA), antisense, RNAi, snRNA, siRNA, miRNA, ta-siRNA, a cosuppression molecule, ribozyme, antibody etc. as described herein, for example under control of an inducible promoter combined with the application of a corresponding inducer, e.g. tetracycline or ecdysone.
  • a modulator such as an antagonist, inhibitor or inductor
  • the reduction, decrease or deletion in activity of the molecule which activity is to be reduced in the process of the invention amounts preferably by at least 10%, preferably by at least 30% or at least 60%, especially preferably by at least 70%, 80%, 85%, 90% or more, very especially preferably are at least 95%, more preferably are at least 99% or more in comparison to the control, reference or wild type. Most preferably the reduction, decrease or deletion in activity amounts to 100%.
  • Various strategies for reducing the quantity, the expression, the activity or the function of proteins encoded by the nucleic acids or the nucleic acid sequences itself according to the invention are encompassed in accordance with the invention. The skilled worker will recognize that a series of different methods are available for influencing the quantity of a protein, the activity or the function in the desired manner.
  • the process of the present invention comprises one or more of the following steps:
  • a protein enabling, mediating or controlling the expression of a protein encoded by the nucleic acid molecule which activity is to be reduced in the process of invention or of the polypeptide which activity is to be reduced in the process of the invention, e.g. of a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7, of Table Il or IV or being encoded by a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7, of Table I;
  • a mRNA molecule enabling, mediating or controlling the expression of a protein to be reduced in the process of the invention or being encoded by the nucleic acid molecule which activity is to be reduced in the process of the invention, e.g. enabling, mediating or controlling the expression of a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7, of Table Il or IV, or of a polypeptide being encoded by a nucleic acid molecule compris- ing a polynucleotide as depicted in Application No.: 1 , column 5 or 7, of Table I,
  • RNA molecule enabling, mediating or controlling the expression of a mRNA encoding a polypeptide which activity is to be reduced in the process of the invention, e.g. of a mRNA encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7, of Table Il or IV, or of a mRNA comprising the nucleic acid molecule which activity is to be reduced in the process of the invention, e.g. comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7, of Table I;
  • RNA molecule enabling, mediating or controlling the expression of an expression product of a nucleic acid molecule comprising the polynucleotide which activity is to be reduced in the process of the invention; e.g. of a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7, of Table I;
  • a mRNA encoding the polynucleotide or the polypeptide which activity is to be reduced in the process of the invention e.g. of a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7, of Table I or of a mRNA enabling, mediating or controlling the expression of a polypeptide which activity is to be reduced in the process of the inven- tion, the polypeptide depicted in Application No.: 1 , column 5 or 7, of Table
  • a gene encoding an activator enabling the activation or increase of the expression of a nucleic acid molecule encoding a polypeptide encoded by the nucleic acid molecule which activity is to be reduced in the process of the invention or the polypeptide which activity is to be reduced in the process of the invention e.g. a gene encoding an activator enabling the activation or increase of the expression of a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted Application No.: 1 , column 5 or 7, of Table Il or IV or of a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7, of Table I; or
  • an endogenous gene encoding the polypeptide or the nucleic acid molecule which activity is to be reduced in the process of the invention for example an endogenous gene encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted Application No.: 1 , column 5 or 7, of Table Il or IV, or a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7, of Table I;
  • RNAi can for example be mediated e.g. by adding or expressing an antisense molecule, co- suppression molecule, an antibody, ribozyme, siRNA, microRNA, ta-siRNA, a cosup- pression molecule, or RNAi, by mutation or deletion of a gene sequence, expressing or improving the activity of a negative expression element or by other methods known to the person skilled in the art or mentioned herein.
  • a polynucleotide which activity is to be reduced in the process of the invention or one or more fragments thereof can for example be expressed in antisense orientation.
  • a hairpin RNAi constructs is expressed. It is also advantageous to express simultaneously a sense and antisense RNA molecule of the nucleic acid molecule or polypeptide which activity is to be reduced in the process of the invention.
  • the present invention relates to a process, wherein the number of functional (e.g. expressed) copies of a gene encoding the polynucleotide or nucleic acid molecule of the invention is decreased.
  • the endogenous level of the polypeptide of the invention can for example be decreased by modifying the transcriptional or translational regulation or effiziency of the polypeptide.
  • the process of the present invention comprises for example one or more of the following steps
  • a)stabilizing a protein conferring the decreased expression of a protein of the nu- cleic acid molecule or polypeptide which activity is to be reduced in the process of the invention b)stabilizing a mRNA or functional RNA conferring the decreased expression of the nucleic acid molecule or polypeptide which activity is to be reduced in the process of the invention; c) increasing or stimulating the specific activity of a protein conferring the decreased expression of the nucleic acid molecule or polypeptide which activity is to be reduced in the process of the invention; d)decreasing the specific activity of a protein conferring the increased expression of the nucleic acid molecule or polypeptide which activity is to be reduced in the process of the invention; e)expressing a transgenic gene encoding a protein conferring the decreased expression of a nucleic acids molecule or polypeptide which activity is to be reduced in the process of the invention, f) generating or increasing the expression of an endogenous or artificial transcrip- tion factor repressing the expression of a protein
  • Lines can be identified in which the repressor elements are integrated near to a gene encoding the nucleic acid molecule or polypeptide which activity is to be reduced in the process of the invention, the expression of which is thereby reduced or deleted.
  • mutations like point mutations can be introduced randomly by different mutagenesis methods and can be selected by specific methods such like TILLING (reviewed in Slade and Knauf, Transgenic Res.
  • an increase of the activity of a protein or RNA leading to a dominant negative phenotype of the protein which activity is to be reduced in the process of the invention can be achieved through the expression of a nucleic acid molecule encoding a protein, which has lost its biological activity but which binds to another protein in a multimeric complex thereby decreasing or deleting the activity of said complex or which binds for example as a transcription factor to DNA and thereby decreasing or deleting the activity of the translated protein.
  • the amount of imRNA, polynucleotide or nucleic acid molecule in a cell or a compartment of an organism correlates to the amount of encoded protein and thus with the overall activity of the encoded protein in said volume. Said correlation is not always linear, the activity in the volume is dependent on the stability of the molecules, the degradation of the molecules or the presence of activating or in- hibiting co-factors. Further, product and educt inhibitions of enzymes are well known.
  • the activity of the abovementioned proteins and/or polypeptide encoded by the nucleic acid molecule to be reduced in the process of the present invention can be reduced, decreased or deleted in various ways.
  • the activity in an organism or in a part thereof, like a cell is reduced or decreased via reducing or decreasing the gene product number, e.g. by reducing or decreasing the expression rate, like mutating the natural promoter to a lower activity, or by reducing or decreasing the stability of the imRNA expressed, thus reducing or de- creasing the translation rate, and/or reducing or decreasing the stability of the gene product, thus increasing the proteins decay.
  • the activity or turnover of enzymes or channels or carriers, transcription factors, and similar active proteins can be influenced in such a manner that a reduction of the reaction rate or a modification (reduction, decrease or deletion) of the affinity to the substrate results, is reached.
  • a mutation in the catalytic centre of a polypeptide or nucleic acid molecule which activity is to be reduced in the process of the invention can modulate the turn over rate of the enzyme, e.g. a knock out of an essential amino acid can lead to a reduced or complete knock out of the activity of the enzyme, or the deletion of regulator binding sites can reduce a positive regulation.
  • the specific activity of an enzyme of the present invention can be decreased such that the turn over rate is decreased or the binding of a co-factor is reduced. Reducing the stability of the encoding imRNA or the protein can also decrease the activity of a gene product. The reduction of the activity is also under the scope of the term "reduced, decreased or deleted activity”. Beside this, advantegously the reduction of the activity in cis, eg. mutating the promotor including other cis-regulatory elements, or the transcribed or coding parts of the gene, inhibition can be achieved in trans, eg.
  • transfactors like chimeric transcription factor, ribozymes, antisense RNAs, dsRNAs or dominant negative proteins versions, which interfere with various stages of expression, eg the transcription, the translation or the activity of the protein or protein complex itself.
  • transfactors like chimeric transcription factor, ribozymes, antisense RNAs, dsRNAs or dominant negative proteins versions, which interfere with various stages of expression, eg the transcription, the translation or the activity of the protein or protein complex itself.
  • epigenetic mechanisms like DNA modifications, DNA methylation, or DNA packaging might be recruited to inactivate or down regulate the nucleic acids of the invention.
  • RNA interference RNA interference
  • dsRNAi RNA interference
  • an antisense nucleic acid RNAi, snRNA, siRNA, imiRNA, ta-siRNA, cosuppression molecule, or a ribozyme nucleic acid combined with an ribozyme
  • a nucleic acid encoding a co-suppressor a nu
  • nucleic acid sequences may be modified so that gene expression is decreased.
  • This reduction, decrease or deletion (reduction, decrease, deletion, inactivation or down-regulation shall be used as synonyms throughout the specification) can be achieved as mentioned above by all methods known to the skilled person, preferably by double-stranded RNA interference (dsRNAi), introduction of an antisense nucleic acid, a ribozyme, an antisense nucleic acid combined with a ribozyme, a nucleic acid encoding a co-suppressor, a nucleic acid encoding a dominant negative protein, DNA- or protein-binding factor or antibodies targeting said gene or -RNA or -proteins, RNA degradation inducing viral nucleic acids and expression systems, systems for inducing a homolog recombination of said genes, mutations in said genes or a combination of the above.
  • dsRNAi double-stranded RNA interference
  • an activity of a gene product in an organism or part thereof, in particular in a plant cell, a plant, or a plant tissue or a part thereof or in a microorganism can be decreased by decreasing the amount of the specific encoding imRNA or the corresponding protein in said organism or part thereof.
  • “Amount of protein or mRNA” is understood as meaning the molecule number of polypeptides or imRNA molecules in an organism, a tissue, a cell or a cell compartment.
  • Decrease in the amount of a protein means the quantitative decrease of the molecule number of said protein in an organism, a tissue, a cell or a cell compartment or part thereof - for example by one of the methods described herein below - in comparison to a wild type, control or reference.
  • activation means that the activity of the polypeptide encoded is essentially no longer detectable in the organism or in the cell such as, for example, within the plant or plant cell.
  • down- regulation means that its activity, e.g. the enzymatic or biological activity of the polypeptide encoded is partly or essentially completely reduced in comparison with the activity of the untreated organism. This can be achieved by different cell- biological mechanisms.
  • the activity can be downregulated in the entire organism or, in the case of multi-celled organisms, in individual parts of the organism, in the case of plants for example in tissues such as the seed, the leaf, the root or other parts.
  • a modification i.e. a decrease
  • a decrease in activity in an organism or a part thereof can be caused by adding a chemical compound such as an antagonist to the media, nutrition, soil of the plants or to the plants themselves.
  • the increase in the fine chemical can be achived by decreasing the level of the endogenous nucleic acid molecule or the endogenous polypeptide described herein, i.e. of the nucleic acid molecule or the polypeptide which activity is to be reduced according to the process of the invention, in particular of a polynucleotide or polypeptide described in in the corresponding line of Table I or II, column 5 or 7, respectively.
  • the reduction or deletion of the activity represented by the protein or nucleic acid molecule to be reduced in the process of the invention is achieved by at least one step selected from the group consisting of:
  • nucleic acid molecule comprising a polynucleotide encoding a ribonucleic acid sequence, which is able to form a double-stranded ribonucleic acid molecule, whereby a fragment of 17 nucleotides (nt) or more, preferably of 18, 19, 20, 21 , 22, 23, 24 or 25 of said double-stranded ribonucleic acid molecule has a homology of 50 % or more, preferably of 60, 65, 70, 75, 80, 85, 90, 95, 97, 98, 99 or most preferred of 100 % to the nucleic acid molecule to be reduced according to the process of the invention or a nucleic acid molecule encoding the polypeptide to be reduced according to the process of the invention or selected from the group consisting of:
  • nucleic acid molecule comprising a polynucleotide as depicted in Application No. 1 , column 5 or 7 of Table I or encoding a polypeptide comprising a polypeptide as depicted in Application No. 1 , column 5 or 7 of Table II, preferably, a nucleic acid molecule as depicted in Application No. 1 , column 5 or 7 of Table I or encoding a polypeptide as depicted in Application No. 1 , column 5 or 7 of Table II, preferably a nucleic acid molecule as depicted in Application No. 1 , column 5 or 7 of Table I A or encoding a polypeptide as depicted in Application No. 1 , column 5 or 7 of Table Il B, and
  • nucleic acid molecule encoding a polypeptide having the activity of poly- peptide depicted in Application No. 1 , column 5 of Table Il or encoding the expression product of a polynucleotide comprising a nucleic acid molecule as depicted in Application No. 1 , column 5 or 7 of Table I;
  • RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, cosup- pression molecule, or an antisense nucleic acid molecule whereby the RNAi, snRNA, dsRNA, siRNA, miRNA, ta-siRNA, cosuppression molecule, or antisense nucleic acid molecule comprises a fragment of 17 nt or more, preferably of 18, 19, 20, 21 , 22, 23, 24 or 25 nt with a identity of at least 30 % or more, preferably of 40, 50, 60, 65, 70, 75, 80, 85, 90, 95, 97, 98, 99 or most preferably of 100 % to the nucleic acid molecule to be reduced according to the process of the invention or a nucleic acid molecule encoding the polypeptide to be reduced according to the process of the invention or to a nucleic acid molecule selected from a group defined in sec- tion (aa) to (a
  • RNAi RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, cosup- pression molecule, ribozyme, antibody, antisense nucleic acid molecule characterized in (b) and the ribozyme characterized in (c);
  • nucleic acid molecule comprising a polynucleotide conferring the expression of a dominant-negative mutant of a protein having the activity of a protein to be reduced according to the process of the invention or of a protein encoded by a nucleic acid molecule to be reduced according to the process of the invention or of a protein encoded by a nucleic acid molecule selected from a group defined in section (aa) to (ac);
  • nucleic acid molecule comprising a polynucleotide encoding a factor, which binds to a nucleic acid molecule comprising the nucleic acid molecule to be reduced according to the process of the invention or comprising a nucleic acid molecule encoding the polypeptide to be reduced according to the process of the invention or comprising a nucleic acid mole- cule selected from a group defined in section (aa) to (ac);
  • nucleic acid construct capable to recombinate with and si- lence, inactivate, repress or reduces the activity of an endogenous gene comprising the nucleic acid molecule to be reduced according to the process of the invention or comprising a nucleic acid molecule encoding the polypeptide to be reduced according to the process of the invention or comprising a nucleic acid molecule selected from a group defined in section (aa) to (ac);
  • the reduction or deletion of the activity represented by the protein or nucleic acid mole- cule used in the process of the invention is achieved by at least one step selected from the group consisting of:
  • nucleic acid molecule to be reduced % to the nucleic acid molecule to be reduced according to the process of the invention or a nucleic acid molecule encoding the polypeptide to be reduced according to the process of the invention or to a nucleic acid molecule selected from the group consisting of:
  • nucleic acid molecule conferring the expression of a protein comprising a polypeptide, a consensus sequence or a polypeptide motif, as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV or conferring the expression of nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7 of Table I;
  • nucleic acid molecule encoding a protein having the activity of a protein to be reduced according to the process of the invention, e.g. comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV or conferring the expression of nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7 of Table I; and
  • nucleic acid molecule comprising a fragment of at least 17, 18, 19, 20, 21 , 22, 23, 24 or 25 base pairs of a nucleic acid molecule with a homology of at least 50% preferably of 60, 65, 70, 75, 80, 85, 90, 95, 97, 98, 99 or 100 % to a nucleic acid molecule of (i) or (ii);
  • nucleic acid molecule antisense nucleic acid molecule has an identity of at least 30% or more, preferably of 40, 50, 60, 65, 70, 75, 80, 85, 90, 95, 97, 98, 99 or 100 % to a nucleic acid molecule antisense to the nucleic acid molecule to be reduced according to the process of the invention or a nucleic acid molecule encoding the polypeptide to be reduced according to the process of the invention or a nucleic acid molecule selected from the group consisting of (i) to (iii) above;
  • a ribozyme which specifically cleaves a nucleic acid molecule conferring the expression of a protein having the activity of a protein to be reduced according to the process of the invention, e.g. comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV, or which specifically cleaves a nucleic acid molecule con- ferring the expression of the nucleic acid molecule to be reduced according to the process of the invention or the polypeptide to be reduced according to the process of the invention or a nucleic acid molecule encoding the polypeptide to be reduced according to the process of the invention or a nucleic acid molecule selected from the group consisting of (i) to (iii) above;
  • nucleic acid molecule conferring the expression of the nucleic acid molecule to be reduced according to the process of the invention or the polypeptide to be reduced according to the process of the invention or a nucleic acid molecule encoding the polypeptide to be reduced according to the process of the invention or a nucleic acid molecule selected from the group consisting of (i) to (iii) above for inducing a co-suppression of the endogenous the nucleic acid molecule to be reduced according to the process of the invention or a nucleic acid molecule encoding the polypeptide to be reduced according to the process of the invention or a nucleic acid molecule selected from the group consisting of
  • nucleic acid molecule conferring the expression of a dominant- negative mutant of a protein having the activity of a protein to be reduced accord- ing to the process of the invention, e.g. comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV or of a dominant-negative mutant of a polypeptide encoded by a nucleic acid molecule selected from the group consisting of (i) to (iii) above, for example expressing said sequence leading to the dominant-negative mutant protein thereby the activity of the protein used in the inventive process is reduced, decreased or deleted and therefore the production of the fine chemical is increased;
  • nucleic acid molecule encoding a factor, which binds to a nucleic acid molecule conferring the expression of a protein having the activity of a polypeptide to be reduced according to the process of the invention, e.g. comprising a polypeptide , a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV or being encoded by a nucleic acid molecule selected from the group consisting of (i) to (iii) above;
  • nucleic acid construct capable to recombinate with and mutate an endogenous gene conferring the expression of a protein having the activity of a protein used in the inventive process especially a polypeptide comprising a polypeptide , a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV or being encoded by a nucleic acid molecule selected from the group consisting of (i) to (iii) above;
  • the process of the present invention comprises the following step:
  • an endogenous nucleic acid molecule e.g. into an endogenous gene, which confers the expression of a polypeptide comprising a polypeptide , a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV or a polypeptide being encoded by a nucleic acid molecule selected from the group consisting of (i) to (iii), a mutation of a distinct amino acid shown in the consensus sequence depicted in column 7 of Table IV under Application 1 in the same line,
  • the mutation confers a non-silent mutation in the polypeptide which activity is to be reduced in the process of the invention, in particular in a polypeptide comprising a polypeptide , a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV or a polypeptide being encoded by a nucleic acid molecule selected from the group consisting of (i) to (iii)
  • the consensus sequence depicted in column 7 of Table IV indicates the amino acids which were found to be strongly conserved within the sequences of the polypeptides depicted in columns 5 and 7 of Application No.: 1 , column 5 or 7 of Table II.
  • the coding sequences of a nucleic acid molecule which activity is to be reduced in the process of the invention in particular from the nucleic acid molecule mentioned under sections (a) to (i) of paragraph [0033.1.1.1], preferably of a nucleic acid molecule comprising a nucleic acid molecule as depicted in Application No.: 1 , column 5 or 7 of Table I, is used for the reduction, decrease or deletion of the nucleic acid sequences which activity is to be reduced in the process of the invention according to the different process steps (a) to (I) mentioned above in para- graphs [0053.1.1.1] to [0054.1.1.1], e.g.
  • bp, 900 bp, 800 bp or 700 bp Preferably less than 600 bp, 500 bp, 400 bp, 300 bp, 200 bp or 100 bp of the coding region of the said nucleic acid sequence are used.
  • nucleic acid sequences disclosed herein as the nucleic acid molecule which activity is to be reduced in the process of the invention to reduce or delete the activity particularly of orthologs of the molecules disclosed herein.
  • the skilled person knows how to isolate the complete gene, the coding region (CDR), the expressed regions (e.g. as cDNA), or fragments thereof of said nucleic acid sequences, in particular said regions of mole- cules as indicated in Table I, column 5 or 7, Application No.: 1 , if not already disclosed herein, e.g.
  • nucleic acid molecule starting from the nucleic acid molecule mentioned under sections (a) to (j) of paragraph [0033.1.1.1] above, preferably starting from a nucleic acid molecule comprising a nucleic acid molecule as depicted in Application No.: 1 , column 5 or 7 of Table I.
  • the 5'- and/or 3'-sequences of a nucleic acid molecule which activity is to be reduced in the process of the invention in particular from the nucleic acid molecule mentioned under sections (a) to (i) of paragraph [0033.1.1.1], preferably of a nucleic acid molecule comprising a nucleic acid molecule as depicted in Application No.: 1 , column 5 or 7 of Table I, is used for the reduction, decrease or dele- tion of the nucleic acid sequences which activity is to be reduced in the process of the invention according to the different process steps (a) to (j) mentioned above in paragraphs [0053.1.1.1] to [0054.1.1.1], e.g.
  • nucleic acid sequences disclosed herein as the nucleic acid molecule which activity is to be reduced in the process of the invention to isolate the UTRs of said molecules.
  • the skilled person knows how to isolate the 5'- and/or 3'-regions of said nucleic acid sequences, in particular the 5'- and/or 3'-regions of the molecules indicated in Table I, column 5 or 7, Application No.: 1 , if not already disclosed herein, e.g.
  • nucleic acid molecule starting from the nucleic acid molecule mentioned under sections (a) to (j) of paragraph [0033.1.1.1] above, prefera- bly starting from a nucleic acid molecule comprising a nucleic acid molecule as depicted in Application No.: 1 , column 5 or 7 of Table I.
  • 5 ' - and 3 ' -regions can be isolated by different methods like RACE (Zang and Frohman (1997) Using rapid amplification of cDNA ends (RACE) to obtain full length cDNAs. Methods MoI Biol 1997;69:61-87 or genomic walking PCR technologies (Mishra et al., 2002, Biotechniques 33(4): 830-832; Spertini et al 1999, Biotechniques 27(2), 308- 314).
  • a reduction in the activity or the function is preferably achieved by a reduced expression of a gene encoding the protein of the inventive process.
  • a polypeptide encoded by nucleic acid molecules comprising the nucleic acid molecules shown in Application No.: 1 , column 5 or 7 of Table I or a polypeptide comprising the amino acid sequences, consensus sequences or polypeptide motifs shown in Application No.: 1 , column 5 or 7 of Table Il or in column 7 of Table IV or a nucleic acid molecule comprising the nucleic acid molecules shown in Application No.: 1 , column 5 or 7 of Table I or encoding a polypeptide comprising the amino acid sequences, consensus sequences or polypeptide motifs shown in Application No.: 1 , column 5 or 7 of Table Il or IV can be achieved for example using the following methods disclosed in paragraph [0059.1.1.1] :
  • dsRNA double-stranded RNA nucleic acid sequence
  • an antisense nucleic acid sequence or of an expression cassette ensuring the expression of the latter.
  • introduction of an antisense nucleic acid sequence or of an expression cassette ensuring the expression of the latter.
  • the antisense nucleic acid sequence is directed against a gene (i.e. genomic DNA sequences including the promoter sequence) or a gene transcript (i.e. RNA sequences) including the 5 ' and 3 ' non-translated regions.
  • RNA sequences i.e. RNA sequences
  • ⁇ -anomeric nucleic acid sequences i.e. genomic DNA sequences including the promoter sequence
  • RNA sequences i.e. RNA sequences
  • RNA or micro-RNA introduction of a microRNA or micro-RNA (imiRNA) that has been designed to target the gene of interest in order to induce a breakdown or translation inhibition of the mRNA of the gene of interest and thereby silence gene expression or of an expression cassette ensuring the expression of the former;
  • [0061.1.1.1] Accordingly, the following paragraphs [0062.1.1.1] to [0555.1.1.1] relate preferably to the repression, reduction or deletion of an activity selected from the group consiting of 2-isopropylmalate synthase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1G13880-protein, At1g23780- protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP-dependent peptidase/ ATPase/ nucleoside- triphosphatase/ serine- type
  • a reference to column 5 or 7, of Table I, Table I A, Table I B, Table II, Table Il A, Table Il B, Table III or Table IV as used herein refers preferably to column 5 or 7 of Application No.: 1 of Table I, Table I A, Table I B, Table II, Table Il A, Table Il B, Table III or Table IV, respectively.
  • RNA nucleic acid sequence e.g. for the reduction or deletion of activity of the nucleic acid molecule or polypeptide which activity is to be reduced in the process of the invention, in particular of a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, consensus sequences or polypeptide motifs as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV
  • dsRNAi double-stranded RNA interference
  • dsRNAi double-stranded RNA interference
  • dsRNAi double-stranded RNA interference
  • dsRNAi double-stranded RNA interference
  • Neurospora Zebrafish
  • Drosophila mice
  • planaria humans
  • Trypanosoma petunia
  • Arabidopsis for example Matzke MA et al. (2000) Plant MoI. Biol. 43: 401-415; Fire A. et al. (1998) Nature 391 : 806-81 1 ; WO 99/32619; WO 99/53050; WO 00/68374; WO 00/44914; WO 00/44895; WO
  • RNAi is also documented as an advantageously tool for the repression of genes in bacteria such as E. coli for example by Tchurikov et al. [J. Biol. Chem., 2000, 275 (34): 26523-26529].
  • Fire et al. named the phenomenon RNAi for RNA interference.
  • the techniques and methods described in the above refer- ences are expressly referred to. Efficient gene suppression can also be observed in the case of transient expression or following transient transformation, for example as the consequence of a biolistic transformation (Schweizer P et al. (2000) Plant J 2000 24: 895-903).
  • dsRNAi methods are based on the phenomenon that the simultaneous intro-duction of complementary strand and counterstrand of a gene transcript brings about highly effective suppression of the expression of the gene in question.
  • the resulting phenotype is very similar to that of an analogous knock-out mutant (Waterhouse PM et al. (1998) Proc. Natl. Acad. Sci. USA 95: 13959-64).
  • RNAi method [0064.1.1.1] Tuschl et al., Gens Dev., 1999, 13 (24): 3191-3197, were able to show that the efficiency of the RNAi method is a function of the length of the duplex, the length of the 3'-end overhangs, and the sequence in these overhangs.
  • dsRNA double-stranded RNA molecule
  • a suitable organism e.g. a microorganism or a plant, or a part thereof - the reduction, re- pression, or deletion of the an activity selected from the group consisting of: 2- isopropylmalate synthase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein,
  • IMS3 2- isopropylmalate synthase
  • the dsRNA molecule of the invention or used in the process of the invention preferable fulfills at least one of the following principles:
  • the 5' and 3' untranslated regions of the used nucleic acid sequence and regions close to the start codon should be in general avoided as this regions are richer in regulatory protein binding sites and interactions between RNAi sequences and such regulatory proteins might lead to undesired interactions; • in plants the 5' and 3' untranslated regions of the used nucleic acid sequence and regions close to the start codon preferably 50 to 100 nt upstream of the start codon give good results and therefore should not be avoided;
  • dsRNA double-stranded RNA sequences from exons are useful for the method, as sequences from introns have no effect;
  • the G/C content in this region should be greater than 30% and less than 70% ideally around 50%;
  • the dsRNAi method can be particularly effective and advantageous for reducing the expression of the nucleic acid molecule which activity is to be reduced in the process of the invention, particular of a nucleic acid molecule comprising a polynu- cleotide depicted in column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, consensus sequences or polypeptide motifs as depicted column 5 or 7 of Table Il or IV and/or homologs thereof.
  • dsRNAi approaches are clearly superior to traditional antisense approaches.
  • the invention therefore furthermore relates to double- stranded RNA molecules (dsRNA molecules) which, when introduced into an organism, advantageously into a plant (or a cell, tissue, organ or seed derived therefrom), bring about altered metabolic activity by the reduction in the expression of the nucleic acid molecule which activity is to be reduced in the process of the invention, particular of a nucleic acid molecule comprising a polynucleotide depicted in Application No.: 1 , col- umn 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, consensus sequences or polypeptide motifs as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV and/or homologs thereof.
  • dsRNA molecules double- stranded RNA molecules
  • RNA molecule of the invention e.g. a dsRNA for reducing the expression of a protein encoded by a nucleic acid molecule which activity is to be re- prised in the process of the invention, particular of a nucleic acid molecule comprising a polynucleotide depicted in Application No.: 1 , column 5 or 7, of Table I and/or homologs thereof,
  • one of the two RNA strands is essentially identical to at least part of a nucleic acid sequence
  • the respective other RNA strand is essentially identical to at least part of the complementary strand of a nucleic acid sequence.
  • the term "essentially identical" refers to the fact that the dsRNA sequence may also include insertions, deletions and individual point mutations in com- parison to the target sequence while still bringing about an effective reduction in expression.
  • the homology as defined above amounts to at least 30%, preferably at least 40%, 50%, 60%, 70% or 80%, very especially preferably at least 90%, most preferably 100%, between the "sense" strand of an inhibitory dsRNA and a part-segment of a nucleic acid sequence of the invention including in a preferred em- bodiment of the invention their endogenous 5 ' - and 3 ' untranslated regions or between the "antisense" strand and the complementary strand of a nucleic acid sequence, respectively.
  • the part-segment amounts to at least 10 bases, preferably at least 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29 or 30 bases, especially preferably at least 40, 50, 60, 70, 80 or 90 bases, very especially preferably at least 100, 200, 300 or 400 bases, most preferably at least 500, 600, 700, 800, 900 or more bases or at least 1000 or 2000 bases or more in length.
  • the part-segment amounts to 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26 or 27 bases, preferably to 20, 21 , 22, 23, 24 or 25 bases. These short sequences are preferred in animals and plants.
  • the longer sequences preferably between 200 and 800 bases are preferred in nonmammalian animals, preferably in invertebrates, in yeast, fungi or bacteria, but they are also useable in plants.
  • an "essentially identical" dsRNA may also be defined as a nucleic acid sequence, which is capable of hy- bridizing with part of a gene transcript (for example in 400 mM NaCI, 40 mM PIPES pH 6.4, 1 mM EDTA at 50°C or 70°C for 12 to 16 h).
  • the dsRNA may consist of one or more strands of polymerized ribonucleotides. Modification of both the sugar-phosphate backbone and of the nucleosides may furthermore be present. For example, the phosphodiester bonds of the natural RNA can be modified in such a way that they encompass at least one nitrogen or sulfur hetero atom. Bases may undergo modification in such a way that the activity of, for example, adenosine deaminase is restricted. These and other modifications are described herein below in the methods for stabilizing antisense RNA.
  • the dsRNA can be prepared enzymatically; it may also be synthesized chemically, either in full or in part. Short dsRNA up to 30 bp, which effectively mediate RNA interference, can be for example efficiently generated by partial digestion of long dsRNA templates using E. coli ribonuclease III (RNase III). (Yang, D., et al. (2002) Proc. Natl. Acad. Sci. USA 99, 9942.) [0070.1.1.1]
  • the double-stranded structure can be formed starting from a single, self- complementary strand or starting from two complementary strands.
  • "sense” and “antisense” sequence can be linked by a linking sequence ("linker”) and form for example a hairpin structure.
  • the linking sequence may take the form of an intron, which is spliced out following dsRNA synthesis.
  • the nucleic acid sequence encoding a dsRNA may contain further elements such as, for example, transcription termination signals or polyadenylation signals. If the two strands of the dsRNA are to be combined in a cell or an organism advantageously in a plant, this can be brought about in a variety of ways:
  • hybridization e.g. crossing of two organisms, advantageously of plants, each of which has been transformed with one vector, one of which encompasses the expression cassette with the "sense” strand while the other encompasses the expression cassette with the "antisense” strand;
  • RNA duplex Formation of the RNA duplex can be initiated either outside the cell or within the cell. If the dsRNA is synthesized outside the target cell or organism it can be introduced into the organism or a cell of the organism by injection, microinjection, electroporation, high velocity particles, by laser beam or mediated by chemical compounds (DEAE-dextran, calciumphosphate, liposomes) or in case of animals it is also possible to feed bacteria such as E. coli strains engineered to express double-stranded RNAi to the animals.
  • chemical compounds DEAE-dextran, calciumphosphate, liposomes
  • the present invention relates to a dsRNA whereby the sense strand of said double-stranded RNA nucleic acid molecule has a homology of at least 30%, 35%, 40%, 45%, 50%, 55% or 60%, preferably 65%, 70%, 75% or 80%, more preferably 85%, 90%, 95%, 96%, 97%, 98% or 99% to the nucleic acid molecule comprising a nucleic acid molecule as shown Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or encoding a polypeptide comprising a polypeptide as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or of Table IV.
  • the encoded sequence or its part- segment of the dsRNA molecule amounts to 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26 or 27 bases, preferably to 20, 21 , 22, 23, 24 or 25 bases, whereby the homology of the sequence is essentially 95%, 96%, 97%, 98%, or preferred 99% or 100%.
  • the expression of the dsRNA molecule of the invention confers the increase of methionine in the organism or part thereof.
  • the sense and antisense strand of the double-stranded RNA are covalently bound or are bound by other, e.g. weak chemical bonds such as hydrogen bonds to each other and the antisense strand is essentially the complement of the sense-RNA strand.
  • the dsRNA may also encompass a hairpin structure, by linking the "sense” and “antisense” strands by a "linker” (for example an intron).
  • a "linker” for example an intron.
  • the self-complementary dsRNA structures are preferred since they merely require the expression of a construct and always encompass the complementary strands in an equimolar ratio.
  • the expression cassettes encoding the "antisense” or the "sense” strand of the dsRNA or the self-complementary strand of the dsRNA are preferably inserted into a vector and stably inserted into the genome of a plant, using the methods described herein below (for example using selection markers), in order to ensure permanent expression of the dsRNA. Transient expression with bacterial or viral vectors are similar useful.
  • the dsRNA can be introduced using an amount which makes possible at least one copy per cell. A larger amount (for example at least 5, 10, 100, 500 or
  • dsRNA which has been generated starting from a nucleic acid molecule to be reduced according to the process of the invention, e.g. of one of the molecules comprising a molecule as shown in column 5 or 7 of Table I or encoding a polypeptide encompassing a polypeptide, a consensus sequence or a motif as shown in column 5 or 7 of Table Il or IV or homologs thereof of the one organism, may be used to suppress the corresponding expression in another organism.
  • dsRNA derived from one of the disclosed nucleic acid molecule to be reduced according to the process of the invention, e.g. of one of the molecules comprising a molecule as shown in column 5 or 7 of Table I or encoding a polypeptide encompassing a polypeptide, a consensus sequence or a polypeptide motif as shown in column 5 or 7 of Table Il or IV or homologs thereof should also have an advantageous effect in other plant species.
  • the dsRNA can be synthesized either in vivo or in vitro.
  • a DNA sequence encoding a dsRNA can be introduced into an expression cassette under the control of at least one genetic control element (such as, for example, pro- moter, enhancer, silencer, splice donor or splice acceptor or polyadenylation signal).
  • at least one genetic control element such as, for example, pro- moter, enhancer, silencer, splice donor or splice acceptor or polyadenylation signal.
  • Suitable advantageous constructs are described herein below. Polyadenylation is not required, nor do elements for initiating translation have to be present.
  • a dsRNA can be synthesized chemically or enzymatically.
  • Cellular RNA polymerases or bacteriophage RNA polymerases (such as, for example T3, T7 or SP6 RNA polymerase) can be used for this purpose.
  • Suitable methods for the in-vitro expression of RNA are described (WO 97/32016; US 5,593,874; US 5,698,425, US 5,712,135, US 5,789,214, US 5,804,693).
  • a dsRNA which has been synthesized in vitro either chemically or enzy- matically can be isolated to a higher or lesser degree from the reaction mixture, for example by extraction, precipitation, electrophoresis, chromatography or combinations of these methods.
  • the dsRNA can be introduced directly into the cell or else be applied extracellularly (for example into the interstitial space).
  • the RNAi method leads to only a partial loss of gene function and therefore en- ables the skilled worker to study a gene dose effect in the desired organism and to fine tune the process of the invention.
  • it leads to a total loss of function and therefore increases the production of the fine chemical. Futher- more it enables a person skilled in the art to study multiple functions of a gene.
  • an antisense nucleic acid sequence e.g. for the reduction or deletion of the nucleic acid molecule or polypeptide which activity is to be reduced in the process of the invention, in particular of a nucleic acid molecule com- prising a polynucleotide depicted in Application No.: 1 , column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV
  • [0081.1.1.1] Methods for suppressing a specific protein by preventing the accumulation of its mRNA by means of "antisense” technology can be used widely and has been described extensively, including for plants; Sheehy et al. (1988) Proc. Natl. Acad. Sci. USA 85: 8805-8809; US 4,801 ,34100; MoI JN et al. (1990) FEBS Lett 268(2): 427- 430.
  • the antisense nucleic acid molecule hybridizes with, or binds to, the cellular mRNA and/or the genomic DNA encoding the target protein to be suppressed. This process suppresses the transcription and/or translation of the target protein.
  • Hybridi- zation can be brought about in the conventional manner via the formation of a stable duplex or, in the case of genomic DNA, by the antisense nucleic acid molecule binding to the duplex of the genomic DNA by specific interaction in the large groove of the DNA helix.
  • an "antisense" nucleic acid molecule comprises a nucleotide sequence, which is at least in part complementary to a "sense" nucleic acid molecule encoding a protein, e.g., complementary to the coding strand of a double- stranded cDNA molecule or complementary to an encoding mRNA sequence. Accordingly, an antisense nucleic acid molecule can bind via hydrogen bonds to a sense nucleic acid molecule.
  • the antisense nucleic acid molecule can be complementary to an entire coding strand of a nucleic acid molecule conferring the expression of the polypeptide to be reduced in the process of the invention or comprising the nucleic acid molecule which activity is to be reduced in the process of the invention or to only a portion thereof. Accordingly, an antisense nucleic acid molecule can be antisense to a "coding region" of the coding strand of a nucleotide sequence of a nucleic acid molecule of the present invention.
  • coding region refers to the region of the nucleotide sequence comprising codons, which are translated into amino acid residues.
  • the antisense nucleic acid molecule is antisense to a "noncod- ing region" of the mRNA flanking the coding region of a nucleotide sequence.
  • noncoding region refers to 5' and 3' sequences which flank the coding region that are not translated into a polypeptide, i.e., also referred to as 5' and 3' untranslated regions (5 ' -UTR or 3 ' -UTR).
  • the noncoding region is in the area of 50 bp, 100 bp, 200bp or 300 bp, peferrably 400 bp, 500 bp, 600 bp, 700 bp, 800 bp, 900 bp or 1000 bp up- and/or downstream from the coding region.
  • antisense nucleic acid molecules Given the coding strand sequences encoding the polypeptide or the nucleic acid molecule to be reduced in the process of the invention, e.g. having above mentioned activity, e.g. the activity of a polypeptide with the activity of the protein which activity is to be reduced in the process of the invention as disclosed herein, antisense nucleic acid molecules can be designed according to the rules of Watson and Crick base pairing.
  • yet another embodiment of the invention is an antisense nucleic acid molecule, which confers - after being expressed in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of the an activity selected from the group consisting of: 2-isopropylmalate synthase
  • IMS3 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1 G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850- protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP-dependent peptidase/ AT- Pase/ nucleoside- triphosphatase/ serine-type endopeptidase, ATR2-protein, beta- galactosidase (BGAL8), calmodulin binding protein / translation elongation factor, choline kinase (ATCK1 ), cytidine deaminase
  • the invention relates to an antisense nucleic acid molecule, whereby the antisense nucleic acid molecule has a homology of at least 30% to a nucleic acid molecule antisense to a nucleic acid molecule encoding the protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or encoding a protein encompassing a consensus sequence or a polypeptide motif as shown in of Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown Application No.: 1 , column 5 or 7 of Table I, preferably as de- picted in Table I B or a homologue thereof as described herein and which confers the increase of the respective fine chemical indicated in the same line of Table I or II, respectively after its expression.
  • the antisense nucleic acid molecule of the invention comprises a fragment of at least 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45 or 50, especially preferably at least 60, 70, 80 or 90 base pairs, very especially preferably at least 100, 200, 300 or 400 base pairs, most preferably at least 500, 600, 700, 800, 900 or more base pairs or at least the entire sequence of a nucleic acid molecule with a homology of at least 50% 60%, 70%, 80% or 90%, preferably 100% to an antisense nucleic acid molecule to a nucleic acid molecule conferring the expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or encoding a protein encompassing a consensus sequence or a polypeptide motif as shown in Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown in Application No.: 1 , column 5
  • An antisense nucleic acid sequence which is suitable for reducing the activity of a protein can be deduced using the nucleic acid sequence encoding this protein, for example the nucleic acid sequence which activity is to be reduced in the proc- ess of the invention, e.g. comprising a nucleic acid molecule as shown in column 5 or 7 of Table I or a nucleic acid molecule encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide as shown in column 5 or 7 of Table Il or IV (or homologs, analogs, paralogs, orthologs thereof), by applying the base-pair rules of Watson and Crick.
  • the antisense nucleic acid sequence can be complementary to all of the transcribed imRNA of the protein; it may be limited to the coding region, or it may only consist of one oligonucleotide, which is complementary to part of the coding or noncoding sequence of the mRNA.
  • the oligonucleotide can be complementary to the nucleic acid region, which encompasses the translation start for the protein.
  • Antisense nucleic acid sequences may have an advantageous length of, for example, 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides but they may also be longer and encompass at least 100, 200, 500, 1000, 2000 or 5000 nucleotides.
  • a particular preferred length is between 15 and 30 nucleotides such as 15, 20, 25 or 30 nucleotides.
  • Antisense nucleic acid sequences can be expressed recombinantly or synthesized chemically or enzymatically using methods known to the skilled worker.
  • an antisense nucleic acid molecule e.g., an antisense oligonucleotide
  • an antisense nucleic acid molecule can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.
  • substances which can be used are phosphorothioate derivatives and acridine-substituted nucleotides such as 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthin, xanthin, 4-acetylcytosine, 5- (carboxyhydroxymethyl)uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxy- methylaminomethyluracil, dihydrouracil, ⁇ -D-galactosylqueosine, inosine, N6- isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7- methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, ⁇ -D- man
  • the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid molecule has been sub- cloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid molecule will be of an antisense orientation to a target nucleic acid molecule of interest, described further in the following subsection).
  • the expression of a protein which activity is to be reduced in the process of the invention e.g. encoded by a nucleic acid molecule comprising a nucleic acid molecule as shown in column 5 or 7 of Table I or of a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as shown in column 5 or 7 of Table Il or IV or homologs, analogs, paralogs, orthologs thereof can be inhibited by nucleotide sequences which are complementary to the regulatory region of a gene (for example a promoter and/or enhancer) and which may form triplex structures with the DNA double helix in this region so that the transcription of the gene is reduced.
  • a nucleic acid molecule comprising a nucleic acid molecule as shown in column 5 or 7 of Table I or of a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as shown in column 5 or 7 of Table Il or IV or homologs, analogs, paralogs, ortholog
  • the antisense nucleic acid molecule can be an ⁇ -anomeric nucleic acid.
  • Such ⁇ -anomeric nucleic acid molecules form specific double- stranded hybrids with complementary RNA in which - as opposed to the conventional ⁇ -nucleic acids - the two strands run in parallel with one another (Gautier C et al. (1987) Nucleic Acids Res. 15: 6625-6641 ).
  • the antisense nucleic acid molecule can also comprise 2'-O-methylribonucleotides (Inoue et al. (1987) Nucleic Acids Res. 15: 6131-6148), or chimeric RNA-DNA analogs (Inoue et al. (1987) FEBS Lett 215: 327-330).
  • the antisense nucleic acid molecules of the invention are typically administered to a cell or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a polypeptide having the activity of protein which activity is to be reduced in the process of the invention or encoding a nucleic acid molecule having the activity of the nucleic acid molecule which activity is to be reduced in the process of the invention and thereby inhibit expression of the protein, e.g., by inhibiting transcription and/or translation and leading to the aforementioned fine chemical increasing activity.
  • the antisense molecule of the present invention comprises also a nu- cleic acid molecule comprising a nucleotide sequences complementary to the regulatory region of an nucleotide sequence encoding the natural occurring polypeptide of the invention, e.g. the polypeptide sequences shown in the sequence listing, or identified according to the methods described herein, e.g., its promoter and/or enhancers, e.g. to form triple helical structures that prevent transcription of the gene in target cells. See generally, Helene, C. (1991 ) Anticancer Drug Des. 6(6):569-84; Helene, C. et al. (1992) Ann. N. Y. Acad. Sci. 660:27-36; and Maher, L.J. (1992) Bioassays 14(12):807-15.
  • Yet another embodiment of the invention is a ribozyme, which confers - after being expressed in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of the activity selected from the group consisting of: 2-isopropylmalate synthase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreduc- tase, anion exchanger, aspartyl protease, AT1 G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP-dependent peptidas
  • the invention relates to a ribozyme, which specifically cleaves a nucleic acid molecule conferring expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or comprising a consensus sequence or a polypeptide motif as depicted in Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or a homologue thereof as described herein, and which confers after its expression the increase of the respective fine chemical methionine, e.g. as indicated in column 6 in the same line of Table I or II, respectively.
  • a ribozyme which specifically cleaves a nucleic acid molecule conferring expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or comprising a consensus sequence or
  • RNA molecules or ribozymes can be adapted to any target RNA and cleave the phosphodiester backbone at specific positions, thus functionally deactivating the target RNA (Tanner NK (1999) FEMS Microbiol. Rev. 23(3): 257-275).
  • the ribozyme per se is not modified thereby, but is capable of cleaving further target RNA molecules in an analogous manner, thus acquiring the properties of an enzyme.
  • ribozyme sequences into "antisense” RNAs imparts this enzyme-like RNA-cleaving property to precisely these "antisense” RNAs and thus increases their efficiency when inactivating the target RNA.
  • the preparation and the use of suitable ribozyme "antisense” RNA molecules is described, for example, by Haseloff et al. (1988) Nature 33410: 585-591.
  • the antisense nucleic acid molecule of the invention can be also a ribozyme.
  • Ribozymes are catalytic RNA molecules with ribonuclease activity, which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region.
  • ribozymes for example "Hammerhead” ribozymes; Haselhoff and Gerlach (1988) Nature 33410: 585-591
  • the ribozyme technology can increase the efficacy of an antisense strategy.
  • yet another embodiment of the invention is a coexpression construct, which confers - after being expressed in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2-isopropylmalate synthase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1G13880- protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP-dependent peptidase/
  • Yet another embodiments of the invention is a coexpression construct conferring the decline or inactivation of a molecule conferring the expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or comprising a consensus sequence or a polypeptide motif as shown in Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or a homo- logue thereof as described herein , e.g. conferring the decline or inactivation of the nu- cleic acid molecule or the polypeptide of the invention, with the result that the level of methionine is increased.
  • the construct introduced may represent the homologous gene to be reduced either in full or only in part.
  • the application of this technique to plants has been described for example by Napoli et al. (1990) The Plant Cell 2: 279-289 and in US 5,03410,323.
  • the above described cosuppression strategy can advantageously be combined with the RNAi method as described by Brummell et al., 2003, Plant J. 33, pp793-800. At least in plants it is advantageously to use strong or very strong promoters in cosuppression approaches.
  • Recent work for example by. Schubert et al., (Plant Journal 2004, 16, 2561-2572 ) has indicated that cosuppression effects are dependent on a gene specific threshold level, above which cosuppression occurs.
  • nucleic acid sequences encoding a dominant-negative protein e.g. for the reduction or deletion of activity of the polypeptide which activity is to be reduced in the process of the invention, in particular of a polypeptide encoded by a nucleic acid molecule comprising a polynucleotide depicted in Application No.: 1 , column 5 or 7, of Table I or of a polypeptide comprising a polypeptide, or a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV.
  • yet another embodiment of the invention is a dominant negative mutant, which confers - after being expressed in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2-isopropylmalate synthase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1 G13880- protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP-dependent peptidase/
  • Yet another embodiment of the invention is a dominate negative mutant conferring the decline or inactivation of a polypeptide conferring the expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or of a polypeptide comprising a consensus sequence or a polypeptide motif as shown in Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or a homologue thereof as described herein , e.g. conferring the decline or inactivation of the nucleic acid molecule or the polypeptide of the invention, with the result that the level of methionine is increased.
  • the function or activity of a protein can efficiently also be reduced by expressing a dominant-negative variant of said protein.
  • the skilled worker is familiar with methods for reducing the function or activity of a protein by means of coexpression of its dominant-negative form [Lagna G and Hemmati-Brivanlou A (1998) Current Topics in Developmental Biology 36: 75-98; Perlmutter RM and Alberola-lla J (1996) Current Opinion in Immunology 8(2): 285-90; Sheppard D (1994) American Journal of Respiratory Cell & Molecular Biology 11 (1 ): 1 -6; Herskowitz I (1987) Nature 329 (6136): 219-22].
  • a dominant-negative variant can be realized for example by changing of an amino acid of a polypeptide encoded by a nucleic acid molecule comprising a polynucleotide depicted in Application No.: 1 , column 5 or 7, of Table I or of a polypep- tide comprising a polypeptide or a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV or homologs thereof.
  • This change can be determined for example by computer-aided comparison ("alignment").
  • Alignment computer-aided comparison
  • These mutations for achieving a dominant-negative variant are preferably carried out at the level of the nucleic acid sequences.
  • a corresponding mutation can be performed for example by PCR-mediated in-vitro mutagenesis using suitable oligonucleotide primers by means of which the desired mutation is introduced.
  • methods are used with which the skilled worker is familiar.
  • the "LA PCR in vitro Mutagenesis Kit” (Takara Shuzo, Kyoto) can be used for this purpose.
  • deleting or changing of functional domains e. g. TF or other signaling components which can bind but not activate may achieve the reduction of protein activity.
  • RNAs or proteins e.g. for the reduction or deletion of activity of the nucleic acid molecule or polypeptide which activity is to be reduced in the process of the invention, in particular of a nucleic acid molecule comprising a polynucleotide depicted in Application No.: 1 , column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide or a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV.
  • yet another embodiment of the invention is a DNA- or protein-binding factor against genes RNAs or proteins, which confers - after being expressed in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2-isopropylmalate syn- thase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CP
  • Yet another embodiment of the invention is a DNA- or protein-binding factor against genes RNAs or proteins conferring the decline or inactivation of a molecule conferring the expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or of a polypeptide comprising a consensus sequence or a polypeptide motif as shown in Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or a homologue thereof as described herein , e.g. conferring the decline or inactivation of the nucleic acid molecule or the polypeptide of the invention, with the result that the level of methionine is increased.
  • a reduction in the expression of a gene encoding the nucleic acid mole- cule or the polypeptide which activity is to be reduced in the process of the invention in particular comprising a nucleic acid molecule comprising a polynucleotide depicted in column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motifas depicted in column 5 or 7 of Table Il or IV or homologs thereof according to the invention can also be achieved with specific DNA- binding factors, for example factors of the zinc finger transcription factor type. These factors attach to the genomic sequence of the endogenous target gene, preferably in the regulatory regions, and bring about repression of the endogenous gene.
  • nucleic acid molecule comprising a polynucleotide depicted in column 5 or 7, of Table I B or encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in column 5 or 7 of Table Il B or homologs thereof, then find the promoter and reduce expression by the use of the abovementioned factors.
  • factors which are introduced into a cell may also be those which themselves inhibit the target protein.
  • the protein-binding factors can, for exam- pie, be aptamers (Famulok M and Mayer G (1999) Curr. Top Microbiol. Immunol. 243: 123-36) or antibodies or antibody fragments or single-chain antibodies. Obtaining these factors has been described, and the skilled worker is familiar therewith.
  • a cytoplasmic scFv antibody has been employed for modulating activity of the phytochrome A protein in genetically modified tobacco plants (Owen M et al. (1992) Biotechnology (NY) 10(7): 790-794; Franken E et al. (1997) Curr. Opin. Biotechnol. 8(4): 41 1-416; Whitelam (1996) Trend Plant Sci. 1 : 286-272).
  • Gene expression may also be suppressed by tailor-made low-molecular-weight synthetic compounds, for example of the polyamide type Dervan PB and B ⁇ rli RW (1999) Current Opinion in Chemical Biology 3: 688-693; Gottesfeld JM et al. (2000) Gene Expr. 9(1-2): 77-91.
  • These oligomers consist of the units 3-(dimethyl- amino)propylamine, N-methyl-3-hydroxypyrrole, N-methylimidazole and N-methyl- pyrroles; they can be adapted to each portion of double-stranded DNA in such a way that they bind sequence-specifically to the large groove and block the expression of the gene sequences located in this position.
  • RNA RNA
  • a nucleic acid molecule comprising a polynu- cleotide depicted in Application No.: 1 , column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV.
  • yet another embodiment of the invention is a viral nucleic acid molecule, which confers - after being expressed in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2-isopropylmalate synthase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1G13880- protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP-dependent peptida
  • Yet another embodiment of the invention is a viral nucleic acid molecule conferring the decline or inactivation of a RNA molecule conferring the expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or a polypeptide comprising a consensus sequence or a polypeptide motif of Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or a homologue thereof as described herein , e.g. conferring the decline or inactivation of the nucleic acid molecule or the polypeptide of the invention, with the result that the level of methionine is increased.
  • Inactivation or downregulation can also be efficiently brought about by inducing specific RNA degradation by the organism, advantageously in the plant, with the aid of a viral expression system (Amplikon) (Angell, SM et al. (1999) Plant J. 20(3): 357-362). Nucleic acid sequences with homology to the transcripts to be suppressed are introduced into the plant by these systems - also referred to as "VIGS” (viral in- Jerusalem gene silencing) with the aid of viral vectors. Then, transcription is switched off, presumably mediated by plant defense mechanisms against viruses. Suitable techniques and methods are described in Ratcliff F et al. (2001 ) Plant J.
  • yet another embodiment of the invention is a construct for inducing a homologous recombination on endogenous genes, which confers - after being introduced in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2- isopropylmalate synthase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1 G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein
  • Yet another embodiment of the invention is a construct for inducing homologous recombination on endogenous genes conferring the decline or inactivation of a molecule conferring the expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or of a polypeptide comprising a consensus sequence or a polypeptide motif as shown in Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or a homologue thereof as described herein , e.g. conferring the decline or inactivation of the nucleic acid molecule or the polypeptide of the invention, with the result that the level of methionine is increased.
  • nucleic acid construct which, for example, comprises at least part of an endogenous gene which is modified by a deletion, addition or substitution of at least one nucleotide in such a way that the functionality is reduced or completely eliminated.
  • the modification may also affect the regulatory elements (for example the promoter) of the gene so that the coding sequence remains unmodified, but expression (transcription and/or translation) does not take place or is reduced.
  • the modified region is flanked at its 5' and 3' end by further nucleic acid sequences, which must be sufficiently long for allowing recombination.
  • Their length is, as a rule, in a range of from one hundred bases up to several kilobases [Thomas KR and Capecchi MR (1987) Cell 51 : 503; Strepp et al. (1998) Proc. Natl. Acad. Sci. USA 95(8): 4368-4373].
  • the host organism - for example a plant - is trans- formed with the recombination construct using the methods described herein below, and clones, which have successfully undergone recombination are selected using for example a resistance to antibiotics or herbicides.
  • the resistance to antibiotics or herbicides can subsequently advantageously be re-eliminated by performing crosses.
  • An example for an efficient homologous recombi- nation system in plants has been published in Nat. Biotechnol. 2002 Oct; 20(10):1030- 4, Terada R et al.: Efficient gene targeting by homologous recombination in rice.
  • Homologous recombination is a relatively rare event in higher eukaryo- tes, especially in plants. Random integrations into the host genome predominate.
  • One possibility of removing the randomly integrated sequences and thus increasing the number of cell clones with a correct homologous recombination is the use of a sequence-specific recombination system as described in US 6,110,736, by means of which unspecifically integrated sequences can be deleted again, which simplifies the selection of events which have integrated successfully via homologous recombination.
  • a multiplicity of sequence-specific recombination systems may be used, examples which may be mentioned being Cre/lox system of bacteriophage P1 , the FLP/FRT system from yeast, the Gin recombinase of phage Mu, the Pin recombinase from E. coli and the R/RS system of the pSR1 plasmid.
  • the bacteriophage P1 Cre/lox system and the yeast FLP/FRT system are preferred.
  • the FLP/FRT and the cre/lox recombinase system have already been applied to plant systems [Odell et al. (1990) MoI. Gen. Genet. 223: 369-378].
  • yet another embodiment of the invention is a mutated homologue of the nucleic acid molecule which activity is to be reduced in the process of the invention and, which confers - after being expressed in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2-isopropylmalate synthase (IMS3), 3- isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1 G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-
  • RNA/DNA oligonucleotides into the plant [Zhu et al. (2000) Nat. Biotechnol. 18(5): 555-558], and the generation of knock-out mutants with the aid of, for example, T-DNA mutagenesis [Koncz et al. (1992) Plant MoI. Biol. 20(5): 963-976], ENU-(N- ethyl-N- nitrosourea) - mutagenesis or homologous recombination [Hohn B and Puchta (1999) H. Proc. Natl. Acad. Sci.
  • Point mutations may also be generated by means of DNA-RNA hybrids also known as "chimeraplasty" [Cole-Strauss et al. (1999) Nucl. Acids Res. 27(5): 1323-1330; Kmiec (1999) Gene Therapy American Scientist 87(3): 240-247].
  • the mutation sites may be specifically targeted or randomly selected. If the mutations have been created randomly e.g. by Transposon-Tagging or chemical mutagenesis, the skilled worked is able to specifically enrich selected muation events in the inventive nucleic acids, especially by different PCR methods know to the person skilled in the art.
  • Mutations can also be introduced by the indroduction of so called homing endonucleases which can be designed to set double strand breaks in specific sequences within the genome. The repair of said double strand breaks often leads to the desired non functional mutations. (Arnould et al (2006) Engineering of large numbers of highly specific homing endonucleases that induce recombination on novel DNA targets. Journal of Molecular Biology. 355(3):443-458).
  • microRNA or micro-RNA
  • a microRNA that has been designed to target the gene of interest in order to induce a breakdown or translational inhibition of the mRNA of the gene of interest and thereby silence gene expression or of an expression cassette ensuring the expression of the former, e.g.
  • nucleic acid molecule for the reduction or deletion of activity of the nucleic acid molecule or polypeptide which activity is to be re- prised in the process of the invention, in particular of a nucleic acid molecule comprising a polynucleotide depicted in Application No.: 1 , column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV.
  • yet another embodiment of the invention is a imiRNA molecule, which con- fers - after being expressed in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2-isopropylmalate synthase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein,
  • a suitable organism e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2-iso
  • Yet another embodiment of the invention is a miRNA molecule conferring the decline or inactivation of a molecule conferring the expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or a polypeptide comprising a consensus sequence or a polypeptide motif as depicted in Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or a homologue thereof as described herein , e.g. conferring the decline or inactivation of the nucleic acid molecule or the polypeptide of the invention, with the result that the level of methionine is increased.
  • MiRNAs have emerged as evolutionarily conserved, RNA- based regulators of gene expression in plants and animals.
  • MiRNAs ⁇ 21 to 25 nt arise from larger precursors with a stem loop structure that are transcribed from non- protein-coding genes.
  • MiRNAs can be efficiently design to specifally target and down regulated selected genes. Determinants of target selection of natural plant imiRNAs have been analysed by Schab and coworkers (Schwab et al. 2005,. 2005 Dev. Cell 8, 517-527). This work has been extended to the design and use of artifical miRNAs (amiRNAs) to efficiently down regulate target genes, resulting in concepts and rules for the design of effective amiRNAs for directed gene silencing
  • ta-siRNA transacting small interfering RNA
  • an expression cassette ensuring the expression of the former.e.g. for the reduction or deletion of activity of the nucleic acid molecule or polypeptide which activity is to be reduced in the process of the invention, in particular of a nucleic acid molecule comprising a polynucleotide depicted in Application No.: 1 , column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypep- tide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV.
  • yet another embodiment of the invention is a ta-siRNA, which confers - after being expressed in a suitable organism, e.g. a microorganism or a plant, or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2-isopropylmalate synthase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1G13880-protein, At1g23780- protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141 -protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP-dependent peptida
  • Yet another embodiment of the invention is a ta-siRNA conferring the decline or inacti- vation of a molecule conferring the expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or a polypeptide comprising a consensus sequence or a polypeptide motif as depicted in Table IV or being encoded by a nucleic acid molecule comprising a polynucleotide as shown in Applica- tion No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or a homo- logue thereof as described herein , e.g. conferring the decline or inactivation of the nucleic acid molecule or the polypeptide of the invention, with the result that the level of methionine is increased.
  • a transacting small interfering RNA can be designed to target the gene of interest in order to induce a breakdown of the imRNA of the gene of interest and thereby silence gene expression.
  • Nucleic acid sequences as described in item B) to K) are expressed in the cell or organism by transformation/transfection of the cell or organism or are introduced in the cell or organism by known methods, for example as disclosed in item A).
  • nucleic acid molecule for the reduction or deletion of activity of the nucleic acid molecule or polypeptide which activity is to be re- prised in the process of the invention, in particular of a nucleic acid molecule comprising a polynucleotide depicted in Application No.: 1 , column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif, as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV.
  • yet another embodiment of the invention is a TILLING or reverse screening primer or a heteroduplex between a mutated DNA and a wild type DNA, which can be used to a identify mutation which confers - after being expressed in a suitable organism, e.g.
  • microorganism or a plant or a part thereof - the reduction, repression, or deletion of an activity selected from the group consisting of: 2-isopropylmalate syn- thase (IMS3), 3-isopropylmalate dehydrogenase / oxidoreductase, anion exchanger, aspartyl protease, AT1G13880-protein, At1g23780-protein, At1g27695-protein, At3g12850-protein, At3g20380-protein, At3g55990-protein, At3g59340-protein, At4g10350-protein, AT4G14713-protein, At4g16141-protein, AT4G20940-protein, At4g34770-protein, AT5G26850-protein, ATP binding protein (CPN60A), ATP- dependent peptidase/ ATPase/ nucleoside- triphosphatase/ serine-type endopeptidase, ATR2-protein, beta
  • Yet another embodiment of the invention is a TILLING or reverse screening primer for identifying a mutation conferring the decline or inactivation of a molecule conferring the expression of a protein as shown in Application No.: 1 , column 5 or 7 of Table II, preferably as depicted in Table Il B, or of a polypeptide comprising a consensus sequence or a polypeptide motif as shown in Table IV or being encoded by a nucleic acid mole- cule comprising a polynucleotide as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, or a homologue thereof as described herein , e.g. conferring the decline or inactivation of the nucleic acid molecule or the polypeptide of the invention, with the result that the level of methionine is increased.
  • a TILLING or a reverse screening primer for the identification of a mutation in a nucleic acid molecule which is a homologue of a nucleic acid molecule shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B, such as a nucleic acid molecule comprising a nucleic acid molecule as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B but which is mutated in one or more nucleotides.
  • the TILLING or reverse screening primer comprises a fragment of at least 17 nucleotides (nt), preferably of 18, 19, 20, 21 , 22, 23, 24, 25, 27, 30 nt. of a nucleic acid molecule shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B.
  • the TILLING or reverse screening primer comprises a fragment of at least 17 nucleotides (nt), preferably of 18, 19, 20, 21 , 22, 23, 24, 25, 27, 30 nt and which is at least 70%, 75%, 80%, 90%, more preferred at least 95%, most preferred 100% homologue to a nucleic acid molecule as shown in Application No.: 1 , column 5 or 7 of Table I, preferably as depicted in Table I B.
  • mutations are induced by treatment with a chemical mutagen (EMS).
  • DNAs are prepared from individuals and arrayed in pools for initial screening. These pools become templates for PCR using primers that amplify a region of interest.
  • Het- eroduplexes are formed between wild-type and mutant fragments in the pool by denaturing and reannealing PCR products. These heteroduplexes are the substrate for cleavage by the nuclease CEL I. After digestion, the resulting products are visualized using standard fluorescent sequencing slab gel electrophoresis. Positive pools are then rescreened as individual DNAs, thus identifying the mutant plant and the approximate position of the mutation along the sequence. This positional information increases the efficiency of sequence analysis, as heterozygous mutations may be otherwise difficult to identify.
  • EMS chemical mutagen
  • High-throughput TILLING is for example described in Colbert et al. (2001 ) Plant Physiology 126: 480-484 and has recently been applied to crops (reviewed in Slade and Knauf, Transgenic Res. 2005 Apr; 14(2): 109-15.
  • organisms are used in which one of the abovementioned genes, or one of the above- mentioned nucleic acids, is mutated in such a manner that the activity of the encoded gene products is influenced by cellular factors to a greater extent than in the reference organism, as compared with the unmutated proteins.
  • This kind of mutation could lead to a change in the metabolic activity of the organism, which than causes in a higher production of the fine chemical.
  • the reason for this higher productivity can be due to a change in regulation mechanism of enzymic activity such as substrate inhibition or feed back regulation.
  • organisms are grown under such conditions, that the expression of the nucleic acids of the invention is reduced or repressed leading to an enhanced production of the fine chemical according to the invention.
  • the amount of the fine chemical in the organism or part thereof can be increase by targeted or random mutagenesis of the endogenous genes comprising or encoding the molecule which activity is to be reduced in the process of the invention, e.g. comprising a polynucleotide as depicted in Application No.; 1 , column 5 or 7 of Table I or encoding an polypeptide comprising a polypeptide, a con- sensus sequence or a polypeptide motif as shown in Application No.: 1 , column 5 or 7 of Table Il or IV.
  • homologous recombination can be used to either introduce negative regulatory elements or to remove, interrupt or delete enhancer elements form regulatory regions.
  • gene conversion like methods described by Ko- chevenko and Willmitzer (Plant Physiol. 2003 May; 132(1 ): 174-84) and citations therein may be modified to disrupt enhancer elements or to enhance to acitivty of negative regulatory elements.
  • mutations or repressing elements can be randomly introduced in (plant) genomes by T-DNA or transposon mutagenesis and lines can be screened for, in which repressing or interrupting elements have be integrated near to a gene of the invention, the expression of which is thereby repressed, reduced or deleted. The inactivation of plant genes by random integrations of enhancer elements has been described.
  • the enhancement of negative regulatory elements or the disruption or weaking of enhancing or activating regulatory elements can also be achieved through common mutagenesis techniques:
  • the production of chemically or radiation mutated populations is a common technique and known to the skilled worker, cf [0551.1.1.1].
  • the expression level can be increased if the endogenous genes encoding a polypeptide or a nucleic acid molecule conferring the activity described herein, in particular genes comprising the nucleic acid molecule of the present invention, are modified by a mutagenesis apporach via homologous recombination with optional identification by TILLING or other reverse screening approaches, or gene conversion.
  • the applicable modification of the nucleic acid molecules described herein for the use in the process of the invention i.e.
  • the reduction, repression or deletion of its acivitiy and being itself encoded by the host organism can for example be achieved by random mutatgenesis with chemicals, radia- tion or UV-light or side directed mutagenesis in such a manner that the production of the fine chemical is increased.
  • This embodiment of the invention shall be deemed as transgenic in the sense of the invention.
  • nucleic acid molecule derived from the polynulceotides described herein for the use in the process of the invention as described herein may be used for the recombinant modification of a wide range of organisms, in particular prokaryotic or eukaryotic microorganisms or plants, so that they become a better and more efficient producer of the fine chemicals due to the deletion or reduction the activity of genes comprising nucleic acid molecule of the invention or of the expression product of said genes according to the process of the invention.
  • the improved production or production efficiency of the fine chemical or products derived therefrom can be brought about by a direct effect of the manipulation or by an indirect effect of this manipulation.
  • nucleic acid molecules disclosed herein or derivates thereof can be incorporated into a nucleic acid construct and/or a vector in such a manner that their introduction into an organism, e.g. a cell, confers an reduced or deleted endogenous or cellulary activity either on the nucleic acid sequence expression level or on the level of the polypeptide encoded by said sequences.
  • nucleic acid molecules encoding the herein disclosed antisense nucleic acid molecule, RNAi, snRNA, dsRNA, siRNA, miRNA, ta- siRNA, cosuppression molecule, ribozyme, antibodies or other molecule inhibiting the expression or activity of an expression product of the nucleic acid molecule to be reduced or deleted in the process of the invention can be incorporated into a nucleic acid construct and/or a vector.
  • the present invention relates to a plant comprising a novel composition of metabolites as well as a novel methionine comprising composition which further comprises other metabolites of the organism.
  • the process of the present invention enables to a novel advantageous amino acids composition, e.g. an extract, from plants or microorganisms comprising a higher content of from a viewpoint of nutrional physiology limited amino acids, like methionine or lysine combined with higher amounts of metabolites positively affecting or lower amounts of metabolites negatively affecting the nutrition or health of animals or humans provided with said compositions or organisms of the invention or parts thereof.
  • a novel advantageous amino acids composition e.g. an extract
  • plants or microorganisms comprising a higher content of from a viewpoint of nutrional physiology limited amino acids, like methionine or lysine combined with higher amounts of metabolites positively affecting or lower amounts of metabolites negatively affecting the nutrition or health of animals or humans provided with said compositions or organisms of the invention or parts thereof.
  • the number or activity of further genes which are required for the import or export of nutrients or metabolites, including amino acids or its precursors, required for the cell's biosynthesis of amino acids may be increased so that the con- centration of necessary or relevant precursors, cofactors or intermediates within the cell(s) or within the corresponding storage compartments is increased.
  • the process according to the invention relates to a process which comprises
  • non-human organism preferably a microorganism, non-human animal, a plant cell, a plant tissue or a plant
  • the organism in particular the microorganism, non-human animal, the plant or animal cell, the plant or animal tissue or the plant is advantageously grown in such a manner that it is not only possible to recover, if desired isolate the free or bound methionine.
  • the organism according to the invention advantageously, a microorganism, a non-human animal, a plant, plant or animal tissue or plant or animal cell, is grown and subse- quently harvested.
  • Examples can be transgenic or non-transgenic plants, cells or protoplasts thereof, transgenic or non-transgenic microorganisms such as fungi, bacteria, yeasts, alga or diatom, cells or protoplasts thereof.
  • Preferred organisms are those which are naturally capable of synthesizing the fine chemical in substantial amounts, like fungi, yeasts, bactria or plants.
  • transgenic animals for example Caenorhabditis elegans, are also suitable as host organisms. Examples of preferred suitable organisms are described in the following paragraphs.
  • Suitable organisms or host organisms for generating the nucleic acid molecule used according to the invention or for the use in the process of the invention to produce the fine chemical, e.g. to be transformed with the nucleic acid construct or the vector (both as described below) of the invention, e.g.
  • RNAi, snRNA, dsRNA, siRNA, imiRNA, ta-siRNA, ribozyme, or antisense molecule or ribozyme or an other molecule inhibiting the expression or activity are, in principle, all organisms which are essentially capable of synthesizing the fine chemical, and which are suitable for the repression, reduction or deletion of genes, in particular of a nucleic acid molecule comprising a polynucleotide as depicted in Application No.: 1 , column 5 or 7, of Table I or encoding a polypeptide comprising a polypeptide, a consensus sequence or a polypeptide motif as depicted in Application No.: 1 , column 5 or 7 of Table Il or IV, .
  • the host or production cells orginate from a microorganism such as a fungus, for example from the genera Aspergillus, Penicillium or Claviceps or from yeasts such as the genera Pichia, Torulopsis, Hansenula, Schizosaccharomyces, Candida, Rhodotorula or Saccharomyces, very especially advantageously from the yeast of the family Saccharomycetaceae, such as the advanta- geous genus Saccharomyces and the very advantageous genus and species Saccharomyces cerevisiae for the production of the fine chemical in microorganims.
  • a microorganism such as a fungus
  • yeasts such as the genera Pichia, Torulopsis, Hansenula, Schizosaccharomyces, Candida, Rhodotorula or Saccharomyces
  • yeast of the family Saccharomycetaceae such as the advanta- geous genus Saccharomy
  • the (transgenic) organism for the production or as source is a microorganism or derived therof, such as an eukaryotic organism, for example a fungus, an alga, diatom or a yeast in particular a fungus, alga, diatom or yeast selected from the families Chaetomiaceae, Choanephoraceae, Cryptococcaceae, Cunninghamell- aceae, Demetiaceae, Moniliaceae, Mortierellaceae, Mucoraceae, Pythiaceae, Sacharo- mycetaceae, Saprolegniaceae, Schizosacharomycetaceae, Sodariaceae, Sporobolo- mycetaceae Tuberculariaceae, Adelotheciaceae, Dinophyceae, Ditrichaceae or Prasi- nophyceae, or a prokaryotic organism, for example a bacterium or blue alga, in particular a bacterium from the families Acti
  • the organism or the cell or the part thereof is a non-human animal such as Caenorhaditis elegans or it is derived thereof
  • Production strains which are especially advantageously selected in the process according to the invention are microorganisms selected from the group of the families Actinomycetaceae, Bacillaceae, Brevibacteriaceae, Corynebacteriaceae, Enterobacteriacae, Gordoniaceae, Micrococcaceae, Mycobacteriaceae, Nocardiaceae, Pseudomonaceae, Rhizobiaceae, Streptomycetaceae, Chaetomiaceae, Choane- phoraceae, Cryptococcaceae, Cunninghamellaceae, Demetiaceae, Moniliaceae, Mor- tierellaceae, Mucoraceae, Pythiaceae, Sacharomycetaceae, Saprolegniaceae, Schizosacharomycetaceae, Sodariaceae, Sporobolomycetaceae, Tuberculariaceae, Adelotheciaceae, Dinophyceae
  • Preferred microorganisms are selected from the group consisting of Chaetomiaceae such as the genera Chaetomium e.g. the species Chaetomidium fimeti; Choan- ephoraceae such as the genera Blakeslea, Choanephora e.g. the species Blakeslea trispora, Choanephora cucurbitarum or Choanephora infundibulifera var. cucurbitarum; Cryptococcaceae such as the genera Candida, Crytococcus, Rhodotorula, Torulopsis e.g.
  • Candida albicans Candida albomarginata, Candida antarctica, Candida bacarum, Candida bogoriensis, Candida boidinii, Candida bovina, Candida brump- tii, Candida cacaoi, Candida cariosilignicola, Candida catenulata, Candida chalmersii, Candida ciferrii, Candida cylindracea, Candida edax, Candida ernobii, Candida famata, Candida freyschussii, Candida friedrichii, Candida glabrata, Candida guillier- mondii, Candida haemulonii, Candida humicola, Candida inconspicua, Candida ingens, Candida intermedia, Candida kefyr, Candida krusei, Candida lactiscondensi, Candida lambica, Candida lipolytica, Candida lusitaniae, Candida macedoniensis, Candida magnoliae, Candida membranaefaciens, Candida mesenterica, Candida multige
  • Cunninghamella blakesleeana Cunning- hamella echinulata, Cunninghamella echinulata var. elegans, Cunninghamella elegans or Cunninghamella homothallica
  • Demetiaceae such as the genera Alternaria, Bipo- laris, Cercospora, Chalara, Cladosporium, Curvularia, Exophilia, Helicosporium, Helminthosporium, Orbimyces, Philalophora, Pithomyces, Spilocaea, Thielaviopsis, Wangiella e.g.
  • Curvularia affinis Curvularia clavata, Curvularia fallax, Curvularia inaequalis, Curvularia indica, Curvularia lunata, Curvularia pallescens, Curvularia verruculosa or Helminothosporium sp.
  • Moniliaceae such as the genera Arthrobot- rys, Aspergillus, Epidermophyton, Geotrichum, Gliocladium, Histoplasma, Micro- sporum, Monilia, Oedocephalum, Oidium, Penicillium, Trichoderma, Trichophyton, Thrichoteclum, Verticillium e.g.
  • Aspergillus aculeatus Aspergillus albus, Aspergillus alliaceus, Aspergillus asperescens , Aspergillus awamori, Aspergillus can- didus, Aspergillus carbonarius, Aspergillus carneus, Aspergillus chevalieri, Aspergillus chevalieri var.
  • Penicillium divaricatum Penicillium diversum,- Penicillium du- clauxii, Penicillium echinosporum, -PBnIcMiUm expansum,-Penicillium fellutanum, Penicillium frequentans,- Penicillium funiculosum,- Penicillium glabrum,- Penicillium gladioli, ⁇ Penicillium griseofulvum,- Penicillium hirsutum,- Penicillium hispanicum, Penicillium islandicum,-Penicillium italicum,-Penicillium italicum var. avellaneum, Penicillium janc- zewskii,-Penicillium janthinellum, -Penicillium japonicum, -Penicillium lavendu-
  • Penicillium lilacinum Penicillium Iividum,-Penicillium martensii, Penicillium mega- sporum,- Penicillium miczynskii,- Penicillium nalgiovense,- Penicillium nigricans, Penicillium notatum,-Penicillium ochrochloron ⁇ Penicillium odoratum,-Penicillium oxalicum Penicillium paraherquei,- Penicillium patulum,- Penicillium pinophilum, Penicillium pis- carium,-Penicillium pseudostromaticum,- Penicillium puberulum ⁇ Penicillium purpuro- genum,-Penicillium raciborskii, -Penicillium roqueforti,- Penicillium rotundum, -Penicillium rubrum,- Penicillium sacculum,- Penicillium
  • Penicillium verrucosum var. cyclopium Penicillium verruculosum
  • Penicillium vinaceum Penicillium violaceum
  • Penicillium viridicatum Penicillium vulpinum
  • Trichoderma hamatum Trichoderma har- zianum
  • Trichoderma koningii Trichoderma Iongibrachiatum
  • Trichoderma polysporum Trichoderma reesei, Trichoderma virens or Trichoderma viride
  • Mortierellaceae such as the genera Mortierella e.g.
  • Mortierella isabellina Mortierella polycephala , Mortierella ramanniana , Mortierella vinacea or Mortierella zonata
  • Mucoraceae such as the genera Actinomucor, Mucor, Phycomyces, Rhizopus, Zygorhynchus e.g. the species Mucor amphibiorum, Mucor circinelloides f. circinelloides, Mucor circinelloides var. griseocyanus, Mucor flavus, Mucor fuscus, Mucor griseocyanus, Mucor heterosporus, Mucor hiemalis, Mucor hiemalis f.
  • Rhizopus microsporus var. rhizopo- diformis Rhizopus nigricans, Rhizopus niveus, Rhizopus oligosporus, Rhizopus oryzae, Rhizopus pygmaeus, Rhizopus rhizopodiformis, Rhizopus semarangensis, Rhizopus thankii, Rhizopus stolonifer, Rhizopus thermosus, Rhizopus tonkinensis, Rhizopus tritici or Rhizopus usamii; Pythiaceae such as the genera Phytium, Phy- tophthora e.g.
  • Saccharomyces ellipsoideus Saccharomyces chevalieri, Saccharomyces delbrueckii, Saccharomyces diastaticus, Saccharomyces drosophilarum, Saccharomyces elegans, Saccharomyces ellipsoideus, Saccharomyces fermentati, Saccharomyces florentinus, Saccharomyces fragilis, Saccharomyces heterogenicus, Saccharomyces hienipiensis, Saccharomyces inusitatus, Saccharomyces italicus, Saccharomyces kluyveri, Saccharomyces krusei, Saccharomyces lactis, Saccharomyces marxianus, Saccharomyces microellipsoides, Saccharomyces montanus, Saccharomyces norbensis, Saccharomyces oleaceus, Saccharomyces paradoxus, Saccharomyces pastorianus, Saccharomyces pretorien
  • Schizosacharomycetaceae such as the genera Schizosaccharomyces e.g. the species Schizosaccharomyces japonicus var. ja- ponicus, Schizosaccharomyces japonicus var. versatilis, Schizosaccharomyces malidevorans, Schizosaccharomyces octosporus, Schizosaccharomyces pombe var. malidevorans or Schizosaccharomyces pombe var. pombe; Sodariaceae such as the genera Neurospora, Sordaria e.g.
  • Tuberculariaceae such as the genera Epi- coccum, Fusarium, Myrothecium, Sphacelia, Starkeyomyces, Tubercularia e.g. the species Fusarium acuminatum, Fusarium anthophilum, Fusarium aquaeductuum, Fusarium aquaeductuum var.
  • Fusarium avenaceum Fusarium buharicum, Fusarium camptoceras, Fusarium cerealis, Fusarium chlamydosporum, Fusarium cilia- turn, Fusarium coccophilum, Fusarium coeruleum, Fusarium concolor, Fusarium crookwellense, Fusarium culmorum, Fusarium dimerum, Fusarium diversisporum, Fusarium equiseti, Fusarium equiseti var.
  • Fusarium moniliforme var. subglutinans Fusarium nivale, Fusarium nivale var. majus, Fusarium oxysporum, Fusarium oxysporum f. sp. aechmeae, Fusarium oxysporum f. sp. cepae, Fusarium oxysporum f. sp. conglutinans, Fusarium oxysporum f. sp. cucumerinum, Fusarium oxysporum f. sp. cyclaminis, Fusarium oxysporum f. sp. dianthi, Fusarium oxysporum f.
  • Fusarium oxysporum f. sp. melonis Fusarium oxysporum f. sp. passi- florae
  • Fusarium oxysporum f. sp. pisi Fusarium oxysporum f. sp. tracheiphilum
  • Fusa- hum oxysporum f. sp. tuberosi Fusarium oxysporum f. sp. tulipae, Fusarium oxysporum f. sp.
  • vasinfectum Fusarium pallidoroseum, Fusarium poae, Fusarium prolif- eratum, Fusarium proliferatum var. minus, Fusarium redolens, Fusarium redolens f. sp. dianthi, Fusarium reticulatum, Fusarium roseum, Fusarium sacchari var. elongatum, Fusarium sambucinum, Fusarium sambucinum var. coeruleum, Fusarium semitectum, Fusarium semitectum var. majus, Fusarium solani, Fusarium solani f. sp.
  • Fusarium sporotrichioides Fusarium sporotrichioides var. minus, Fusarium sublunatum, Fusarium succisae, Fusarium sulphureum, Fusarium tabacinum, Fusarium tricinctum, Fusarium udum, Fusarium ventricosum, Fusarium verticillioides, Fusarium xylarioides or Fusarium zonatum; Sporobolomycetaceae such as the genera Bullera, Sporobolomy- ces, ltersonilia e.g.
  • Ditrichaceae such as the genera Ceratodon, Pleuridium, Astomiopsis, Ditrichum, Philibertiella, Cera- todon, Distichium, Skottsbergia e.g. the species Ceratodon antarcticus, Ceratodon pur- pureus, Ceratodon purpureus ssp. convolutes or Ceratodon purpureus ssp.
  • Prasinophyceae such as the genera Nephroselmis, Prasinococcus, Scherffelia, Tetraselmis, Mantoniella, Ostreococcus e.g. the species Nephroselmis olivacea, Prasi- nococcus capsulatus, Scherffelia dubia, Tetraselmis chui, Tetraselmis suecica, Mantoniella squamata or Ostreococcus tauri; Actinomycetaceae such as the genera Actinomyces, Actinobaculum, Arcanobacterium, Mobiluncus e.g.
  • curtisii Mobiluncus curtisii subsp. holmesii or Mobiluncus mulieris
  • Bacillaceae such as the genera Amphibacillus, Anoxybacillus, Bacillus, Ex- iguobacterium, Gracilibacillus, Holobacillus, Saccharococcus, Salibacillus, Virgibacillus e.g.
  • Corynebacterium accolens, Corynebacterium afermentans subsp. afermentans, Corynebacterium afermentans subsp. lipophilum, Corynebacterium ammoniagenes, Corynebacterium amycolatum, Corynebacterium appendicis, Corynebacterium aquilae, Corynebacterium argentoratense, Corynebacterium atypicum, Corynebacterium auri- mucosum, Corynebacterium auris, Corynebacterium auriscanis, Corynebacterium be- tae, Corynebacterium beticola, Corynebacterium bovis, Corynebacterium callunae, Corynebacterium camporealensis, Corynebacterium capitovis, Corynebacterium casei, Corynebacterium confusum, Corynebacterium co
  • Salmonella choleraesuis subsp. bongori Salmonella choleraesuis subsp. cholereasuis, Salmonella choleraesuis subsp. diarizonae, Salmonella choleraesuis subsp. houtenae, Salmonella choleraesuis subsp. indica, Salmonella choleraesuis subsp. salamae, Salmonella daressalaam, Salmonella enterica subsp. houtenae, Salmonella enterica subsp.
  • Gordoniaceae such as the genera Gordonia, Sker- mania e.g. the species Gordonia aichiensis, Gordonia alkanivorans, Gordonia amarae, Gordonia amicalis, Gordonia bronchialis, Gordonia desulfuricans, Gordonia hirsuta, Gordonia hydrophobica, Gordonia namibiensis, Gordonia nitida, Gordonia paraf- finivorans, Gordonia polyisoprenivorans, Gordonia rhizosphera, Gordonia rubriper- tincta, Gordonia sihwensis, Gordonia sinesedis, Gordonia sputi, Gordonia terrae or Gordonia westfalica; Micrococcaceae such as the genera Micrococcus, Arthrobacter, Kocuria, Nesterenkonia, Renibacterium, Rothia, Stomatococc
  • Micrococcus agilis the species Micrococcus agilis, Micrococcus antarcticus, Micrococcus halobius, Micrococcus kris- tinae, Micrococcus luteus, Micrococcus lylae, Micrococcus nishinomiyaensis, Micrococcus roseus, Micrococcus sedentarius, Micrococcus varians, Arthrobacter agilis, Arthrobacter albus, Arthrobacter atrocyaneus, Arthrobacter aurescens, Arthrobacter chlorophenolicus, Arthrobacter citreus, Arthrobacter creatinolyticus, Arthrobacter crys- tallopoietes, Arthrobacter cumminsii, Arthrobacter duodecadis, Arthrobacter flavescens, Arthrobacter flavus, Arthrobacter gandavensis, Arthrobacter globiformis, Arthrobacter histidinolovorans, Arthrobacter ilicis,
  • Mycobacterium africanum the species Mycobacterium africanum, Mycobacterium agri, Mycobacterium aichiense, Mycobacterium alvei, Mycobacterium asiaticum, Mycobacterium aurum, Mycobacterium austroafricanum, Mycobacterium bohemicum, Myco- bacterium botniense, Mycobacterium brumae, Mycobacterium chelonae subsp.
  • pseudoalcali- genes Pseudomonas pseudoflava, Pseudomonas putida, Pseudomonas putida var. naraensis, Pseudomonas putrefaciens, Pseudomonas pyrrocinia, Pseudomonas ra- diora, Pseudomonas reptilivora, Pseudomonas rhodesiae, Pseudomonas rhodos, Pseudomonas riboflavina, Pseudomonas rubescens, Pseudomonas rubrisubalbicans, Pseudomonas ruhlandii, Pseudomonas saccharophila, Pseudomonas savastanoi, Pseudomonas savastanoi pvar.
  • coronafaciens Pseudomonas syringae pvar. delphinii, Pseudomonas syringae pvar. glycinea, Pseudomonas syringae pvar. helianthi, Pseudomonas syringae pvar. lachrymans, Pseudomonas syringae pvar. lapsa, Pseudomonas syringae pvar. morsprunorum, Pseudomonas syringae pvar. phaseolicola, Pseudomonas syringae pvar.

Landscapes

  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
EP07857190A 2006-04-05 2007-04-05 Verfahren zur herstellung einer feinen chemikalie Withdrawn EP1991689A1 (de)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP09156643A EP2090662A3 (de) 2006-04-05 2007-04-04 Verfahren zur Herstellung einer feinen Chemikalie
EP07857190A EP1991689A1 (de) 2006-04-05 2007-04-05 Verfahren zur herstellung einer feinen chemikalie

Applications Claiming Priority (52)

Application Number Priority Date Filing Date Title
EP06112485 2006-04-05
EP06112495 2006-04-07
EP06112737 2006-04-12
EP06114210 2006-05-15
EP06114273 2006-05-18
EP06114252 2006-05-18
EP06114258 2006-05-18
EP06114677 2006-05-19
EP06117394 2006-05-24
EP06115524 2006-06-13
EP06115416 2006-06-14
EP06116792 2006-07-07
EP06116788 2006-07-07
EP06116881 2006-07-10
EP06117578 2006-07-20
EP06117985 2006-07-27
EP06118026 2006-07-28
EP06118188 2006-07-31
EP06118329 2006-08-02
EP06118398 2006-08-03
EP06118424 2006-08-04
EP06118500 2006-08-07
EP06118634 2006-08-09
EP06118776 2006-08-11
EP06119016 2006-08-16
EP06119134 2006-08-18
EP06119211 2006-08-21
EP06119448 2006-08-24
EP06119512 2006-08-25
EP06119747 2006-08-30
EP06120256 2006-09-07
EP06120634 2006-09-14
EP06120898 2006-09-19
EP06121097 2006-09-22
EP06121250 2006-09-26
EP06121334 2006-09-27
EP06121485 2006-09-29
EP06121594 2006-10-02
EP06121602 2006-10-02
EP06123065 2006-10-27
EP07100082 2007-01-04
EP07100145 2007-01-05
EP07100403 2007-01-11
EP07100640 2007-01-17
EP07100643 2007-01-17
EP07100782 2007-01-19
EP07100934 2007-01-22
EP07100951 2007-01-23
EP07100952 2007-01-23
EP07101147 2007-01-25
PCT/EP2007/053344 WO2008034648A1 (en) 2006-04-05 2007-04-04 Process for the production of a fine chemical
EP07857190A EP1991689A1 (de) 2006-04-05 2007-04-05 Verfahren zur herstellung einer feinen chemikalie

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP09156643A Division EP2090662A3 (de) 2006-04-05 2007-04-04 Verfahren zur Herstellung einer feinen Chemikalie

Publications (1)

Publication Number Publication Date
EP1991689A1 true EP1991689A1 (de) 2008-11-19

Family

ID=39869950

Family Applications (1)

Application Number Title Priority Date Filing Date
EP07857190A Withdrawn EP1991689A1 (de) 2006-04-05 2007-04-05 Verfahren zur herstellung einer feinen chemikalie

Country Status (1)

Country Link
EP (1) EP1991689A1 (de)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10760091B2 (en) 2007-06-06 2020-09-01 Monsanto Technology Llc Genes and uses for plant enhancement

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2008034648A1 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10760091B2 (en) 2007-06-06 2020-09-01 Monsanto Technology Llc Genes and uses for plant enhancement
US11371054B2 (en) 2007-06-06 2022-06-28 Monsanto Technology Llc Genes and uses for plant enhancement

Similar Documents

Publication Publication Date Title
WO2008034648A1 (en) Process for the production of a fine chemical
US8952217B2 (en) Process for decreasing verbascose in a plant by expression of a chloroplast-targeted fimD protein
EP2096177A2 (de) Verfahren zur Herstellung von Lutein
US20140325709A1 (en) Process for the production of fine chemicals
US20140259212A1 (en) Process for the Production of Fine Chemicals
US20140199313A1 (en) Process for the Production of Fine Chemicals
CN101675069A (zh) 产生精细化学品的方法
CN106222166B (zh) 用于在植物中增强种子特异性基因表达而促进增强的多不饱和脂肪酸合成的调节性核酸分子
CN101421412A (zh) 控制精细化学品产生的方法
CN107532179B (zh) 产生pufa的材料和方法及含有pufa的组合物
EP1953235A2 (de) Neue Gene im Zusammenhang mit einem Produktionsverfahren für Feinchemikalien
US20120005777A1 (en) Process for the Production of Fine Chemicals
CN102597242B (zh) 新脂肪酸延伸组合物及其用途
WO2001044276A2 (en) Moss genes from physcomitrella patens encoding proteins involved in the synthesis of tocopherols carotenoids and aromatic amino acids
EP2368905A2 (de) Verfahren zur Steuerung der Herstellung von Feinchemikalien
AU2013200435A1 (en) Process for the production of fine chemicals
EP1991689A1 (de) Verfahren zur herstellung einer feinen chemikalie
MXPA06014967A (es) Proceso para laproduccion de productos quimicos finos
WO2020049155A1 (en) Improved method for the production of high levels of pufa in plants
AU2011244938A1 (en) Process for the production of fine chemicals

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080812

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

17Q First examination report despatched

Effective date: 20111108

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20140827