(19)
(11)EP 3 380 627 B1

(12)EUROPEAN PATENT SPECIFICATION

(45)Mention of the grant of the patent:
14.08.2019 Bulletin 2019/33

(21)Application number: 16790964.7

(22)Date of filing:  01.11.2016
(51)International Patent Classification (IPC): 
C12P 13/12(2006.01)
(86)International application number:
PCT/EP2016/076281
(87)International publication number:
WO 2017/089077 (01.06.2017 Gazette  2017/22)

(54)

METHOD FOR PRODUCING L-METHIONINE

VERFAHREN ZUR HERSTELLUNG VON L-METHIONIN

PROCÉDÉ DE FABRICATION DE L- MÉTHIONINE


(84)Designated Contracting States:
AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

(30)Priority: 27.11.2015 EP 15196776

(43)Date of publication of application:
03.10.2018 Bulletin 2018/40

(73)Proprietor: Evonik Degussa GmbH
45128 Essen (DE)

(72)Inventors:
  • OCHROMBEL, Ines
    33729 Bielefeld (DE)
  • FISCHER, Daniel
    Midlothian, Virginia 23113 (US)
  • BATHE, Brigitte
    33154 Salzkotten (DE)
  • HASSELMEYER, Marleen
    33098 Paderborn (DE)
  • HAMPEL, Michael
    39359 Velsdorf (DE)
  • PEDALL, Joanne
    49326 Melle (DE)

(74)Representative: Evonik Patent Association 
c/o Evonik Industries AG IP Management Bau 1042A/PB 15 Paul-Baumann-Straße 1
45772 Marl
45772 Marl (DE)


(56)References cited: : 
EP-A2- 2 292 783
WO-A2-2007/011939
  
  • BOLTEN CHRISTOPH J ET AL: "Towards Methionine Overproduction in Corynebacterium glutamicum - Methanethiol and Dimethyldisulfide as Reduced Sulfur Sources", JOURNAL OF MICROBIOLOGY AND BIOTECHNOLOGY, vol. 20, no. 8, August 2010 (2010-08), pages 1196-1203, XP002754158, cited in the application
  • KOSUGE T ET AL: "ANALYSIS OF THE METHIONINE BIOSYNTHETIC PATHWAY IN THE EXTREMELY THERMOPHILIC EUBACTERIUM THERMUS THERMOPHILUS", JOURNAL OF BIOSCIENCE AND BIOENGINEERING, ELSEVIER, AMSTERDAM, NL, vol. 90, no. 3, 1 January 2000 (2000-01-01), pages 271-279, XP001037455, ISSN: 1389-1723
  
Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


Description


[0001] The present invention relates to a method for producing L-methionine, wherein a microorganism having L-homoserine O-acetyltransferase activity and O-acetyl-L-homoserine sulfhydrylase activity is cultured in the presence of L-homoserine and methyl mercaptan, a salt of the same or dimethyl disulfide whereby the L-methionine is accumulated in the culture medium.

[0002] The amino acid methionine is currently industrially produced worldwide in large amounts and is of considerable commercial importance. Methionine is employed in many fields, such as pharmaceutical, health and fitness products, but particularly as feedstuff additive in many feedstuffs for various livestock, where both the racemic and the enantiomerically pure form of methionine may be used.

[0003] On an industrial scale, methionine is produced chemically via the Bucherer-Bergs reaction, which is a variant of the Strecker synthesis. In this case, the starting substances methylmercaptopropionaldehyde (prepared from acrolein and methyl mercaptan), hydrogen cyanide, ammonia and carbon dioxide are reacted to give 5-(2-methylmercaptoethyl) hydantoin (methionine hydantoin), this subsequently being hydrolysed by alkali to give the alkali metal methioninate and the methionine then being liberated by neutralisation with acid (EP 0 780 370 A2). Various other methods can also be used to prepare methionine, for example, the amidocarbonylation reaction, the hydrolysis of proteins or the fermentation of microorganisms producing-methionine. In chemical synthesis, methionine is produced as a racemic mixture of D- and L-methionine, whereas L-methionine, or L-configured precursors of the same, L-homoserine for example, can be produced by the fermentation of suitable microorganisms.

[0004] L-Homoserine, a potential precursor of L-methionine (H.J. Teas et al., J. Biol. Chem. 1948, 172: 651-658), can be produced both chemically (M.D. Armstrong, J. Am. Chem. Soc., Vol. 70, 1756-1759, 1948) and by fermentation by means of microorganisms (see e.g. US 3,598,701, US 6,303,348 B1, EP 0 994 190 A2, EP 1 149 911 A2, WO 2004/067757 A1).

[0005] Hateley et al. disclose a method in which L-methionine is obtained by a chemical route starting from L-homoserine (WO 2007/085514 A2).

[0006] Lievense was able to prove that microorganism strains lacking homocysteine methylase activity, whose L-homoserine production had been upregulated and which had been transformed with a plasmid encoding an L-homoserine acetyltransferase and an O-acetyl-L-homoserine sulfhydrylase (O-acetylhomoserine (thiol)-lyase), produced L-methionine in excess of their own need in the presence of methyl mercaptan, in contrast to the original strain (E. coli, C. glutamicum or B. flavum) (WO 93/17112 A1).

[0007] Bolten et al. (J. Microbiol. Biotechnol. (2010), 20(8), 1196-1203) could show that (wildtype) C. glutamicum is able to grow on methyl mercaptan and on its dimeric form, dimethyl disulfide, as sole sulphur sources instead of sulfate, the most common sulphur source for the cultivation of microorganisms, and investigated the underlying pathways and enzymes. They demonstrated that MetY (O-acetyl-L-homoserine sulfhydrylase) is responsible for the substitution of the acetyl group of O-acetyl-L-homoserine by the mercapto group of methylmercaptan or of dimethyl disulfide to directly yield L-methionine. In order to increase L-methionine production the authors suggest the amplification not only of the MetY, but also of other enzymes of the L-methionine biosynthesis.

[0008] Zelder et al. (WO 2007/011939 A2) demonstrated, that L-methionine can be produced in microorganisms, such as E. coli and C. glutamicum, by culturing the microorganisms having a deregulated O-acetyl-L-homoserine sulfhydrylase or O-succinyl-L-homoserine sulfhydrylase and/or L-homoserine acetyltransferase or L-homoserine succinyltransferase in the presence of a methyl capped sulfide compound, such as dimethyl disulfide or dimethyl trisulfide.

[0009] A two-stage biotechnological method for preparing L-methionine is proposed by Kim et al. (WO 2008/013432 A1). In a first step here, an L-methionine precursor, O-succinyl-L-homoserine or O-acetyl-L-homoserine, is initially obtained by means of recombinant microorganisms, which are accumulated in the culture broth. In the subsequent second step, the L-methionine precursor is reacted with methyl mercaptan in the presence of a protein having O-succinyl-L-homoserine sulfhydrylase activity or O-acetyl-L-homoserine sulfhydrylase activity or in the presence of a microorganism producing such protein or a cell digest of this microorganism, to give L-methionine and the corresponding carboxylic acid, i.e. acetate or succinate.

[0010] However, in this enzymatic reaction, equimolar amounts of acetate or succinate are formed in addition to L-methionine. On choosing O-acetyl-L-homoserine as L-methionine precursor, for example, this leads to high acetate concentrations in the course of the reaction, particularly on an industrial scale. At a low external pH, undissociated acetate molecules can get into the cell across the membrane and be deprotonated therein, which leads to a fall in the internal pH of the cytoplasm and which perturbs the cell pH homeostasis (I.R. Booth, Microbiological Reviews 49, No. 4 (1985), 359-378). Furthermore, acetate cannot be completely removed from the L-methionine product with acceptable effort. Accordingly, Hong et al. (WO 2012/091479 A2) propose numerous methods to remove and to reuse the relatively large amounts of acetate generated in the second stage of the L-methionine production process from the L-methionine product.

[0011] The object of the present invention is to provide a process for producing L-methionine in a microorganism in which the acetate formed in the conversion of O-acetyl-L-homoserine to L-methionine is substantially reused by the same microorganism.

[0012] This object is achieved by a method for producing L-methionine, wherein a microorganism having L-homoserine O-acetyltransferase activity and O-acetyl-L-homoserine sulfhydrylase activity is cultured in a culture medium comprising L-homoserine and a sulphur source, the sulphur source being selected from the group consisting of methyl mercaptan (MC), a methyl mercaptan salt and dimethyl disulfide (DMDS), whereby L-methionine is accumulated in the culture medium

[0013] Enzyme activities in microorganisms are generally effected by the expression of the corresponding gene encoding the respective enzyme. So-called promoters are located upstream of the gene. A promoter is a DNA sequence consisting of about 40 to 50 base pairs and which constitutes the binding site for an RNA polymerase holoenzyme and the transcriptional start point (M. Pátek et al., Microbial Biotechnology, 6 (2013), 103-117), whereby the strength of expression of the controlled polynucleotide or gene can be influenced. A "functional linkage" is understood to mean the sequential arrangement of a promoter with a gene, which leads to a transcription of the gene.

[0014] The microorganism may also be recombinant and have enhanced L-homoserine O-acetyltransferase activity and enhanced O-acetyl-L-homoserine sulfhydrylase activity.

[0015] Enhanced enzyme activities in microorganisms can be effected, for example, by mutation of the corresponding endogenous gene. Enzyme activities can also be enhanced by increasing the expression of the corresponding gene, for example, by increasing the gene copy number and/or by enhancing gene regulatory factors. The enhancement of such regulatory factors which positively influence gene expression can, for example, be achieved by modifying the promoter sequence upstream of the structural gene in order to increase the effectiveness of the promoter or by completely replacing said promoter with a more effective promoter.

[0016] In the method according to the present invention, the L-homoserine O-acetyltransferase activity and the O-acetyl-L-homoserine sulfhydrylase activity are both preferably enhanced by increased expression of a gene which encodes a protein having L-homoserine O-acetyltransferase activity or a protein having O-acetyl-L-homoserine sulfhydrylase activity. The increased gene expression is preferably achieved by increasing the copy number of the gene encoding the protein having L-homoserine O-acetyltransferase activity or the protein having O-acetyl-L-homoserine sulfhydrylase activity and/or by functional linkage in each case of the gene encoding the protein having L-homoserine O-acetyltransferase activity or the protein having O-acetyl-L-homoserine sulfhydrylase activity to a strong promoter.

[0017] Suitable strong promoters or methods of producing such promoters for increasing expression are known from the literature (e.g. S. Lisser and H. Margalit, Nucleic Acid Research, 1993, Vol. 21, No. 7, 1507-1516; M. Pátek and J. Nesvera in H. Yukawa and M Inui (eds.), Corynebacterium glutamicum, Microbiology Monographs 23, Springer Verlag Berlin Heidelberg 2013, 51-88; B. J. Eikmanns et al., Gene, 102 (1991) 93-98). For instance, native promoters may be optimized by altering the promoter sequence in the direction of known consensus sequences with respect to increasing the expression of the genes functionally linked to these promoters (M. Patek et al., Microbiology (1996), 142, 1297-1309; M. Patek et al., Microbial Biotechnology 6 (2013), 103-117).

[0018] To increase the expression of the gene encoding the protein having L-homoserine O-acetyltransferase activity (metX) or the gene encoding the protein having O-acetyl-L-homoserine sulfhydrylase activity (metY), the tacl promoter (Ptacl), for example, is suitable (H.A. deBoer et al., Proc. Natl. Acad. Sci. USA, Vol. 80, 21-25, January 1983, Biochemistry). The sequence of Ptacl is shown under sequence number 5 (SEQ ID No. 5).

[0019] Constitutive promoters are also suitable for the overexpression, in which the gene encoding the enzyme activity is expressed continuously under the control of the promoter such as, for example, the glucose dependent deo promoter. Chemically induced promoters are also suitable, such as tac, lac or trp. The most widespread system for the induction of promoters is the lac operon of E. coli. In this case, either lactose or isopropyl β-D-thiogalactopyranoside (IPTG) is used as inducer. Also systems using arabinose (e.g. the pBAD system) or rhamnose (e.g. E. coli KRX) are common as inducers. A system for physical induction is, for example, the temperature-induced cold shock promoter system based on the E. coli cspA promoter from Takara or Lambda PL and also osmotically inducible promoters, for example, osmB (e.g. WO 95/25785 A1).

[0020] In the method according to the present invention, the recombinant microorganism is selected from the group consisting of Enterobacteriaceae and Corynebacteriaceae, for example an Escherichia coli (E. coli) strain, e.g. the non-pathogenic E. coli K-12 strain MG1655 (DSM 18039), or a Corynebacterium glutamicum (C. glutamicum) strain, e.g. ATCC13032, or a Corynebacterium humireducens (C. humireducens) strain, e.g. DSM 45392.

[0021] In the method according to the present invention, the L-homoserine O-acetyltransferase activity is, for example, the enzyme MetX, which originates from Corynebacterium glutamicum. The enzyme MetX may also originate from C.

[0022] humireducens. Kim et al. (EP 2 657 345 A1; EP 2 657 250 A2) or Ochrombel et al. (WO 2015/165746 A1) disclose examples of suitable enzymes having L-homoserine O-acetyltransferase activity. The enzyme MetX used in the experimental examples described below has the amino acid sequence according to sequence number 2 (SEQ ID No. 2). The corresponding nucleotide sequence for the gene metX is shown under sequence number 1 (SEQ ID No. 1). The sequence originates from C. glutamicum (ATTC13032) NC_003450.

[0023] An O-acetyl-L-homoserine sulfhydrylase activity suitable for the method according to the present invention is, for example, the enzyme MetY, which originates from Corynebacterium glutamicum. The enzyme MetY may also originate from C. humireducens. Möckel et al. (WO 02/18613 A1), Kröger et al. (WO 2007/024933 A2) or Kim et al. (EP 2 657 345 A1) disclose examples of enzymes having O-acetyl-L-homoserine sulfhydrylase activity according to the invention. The enzyme MetY used in the experimental examples described below has the amino acid sequence according to sequence number 4 (SEQ ID No. 4). The

[0024] corresponding nucleotide sequence for the gene metY is shown under sequence number 3 (SEQ ID No. 3). The sequence originates from C. glutamicum (ATTC13032) NC_003450.

[0025] L-Homoserine is transported into the microorganisms via importers for branched-chain amino acids, e.g. the LIV system in Escherichia coli (B.A. Templeton and M. A. Savageau, JOURNAL OF BACTERIOLOGY, Vol. 117, No. 3, Mar. 1974, p. 1002-1009). In Corynebacterium glutamicum (C. glutamicum) there is also an homologous transport system encoded by, BrnQ of cgl2310 (A. Tauch et al., Arch Microbiol 169 (1998): 303-312).

[0026] Within the cell, the L-homoserine is activated on its hydroxyl group by the transfer of the acetyl group of acetyl coenzyme A (acetyl-CoA) to give O-acetyl-L-homoserine via an (heterologous) homoserine O-acetyltransferase (MetX). The O-acetyl-L-homoserine is then converted to L-methionine and acetate in the presence of a reduced sulphur source, such as methyl mercaptan (MC), and of pyridoxal 5'-phosphate (PLP) by an (heterologous) sulfhydrylase (MetY). Whereas O-acetyl-L-homoserine is one of the natural intermediates of methionine biosynthesis in corynebacteria, methionine biosynthesis in enterobacteria proceeds analogously via an O-succinyl-L-homoserine intermediate (see e.g. Figge R (2007) Methionine biosynthesis in Escherichia coli and Corynebacterium glutamicum. In: Wendisch VF (ed) Amino acid biosynthesis - pathways, regulation and metabolic engineering. Microbiology Monographs, vol 5. Springer, Berlin, pp 163-193). Therefore, the L-homoserine O-acetyltransferase and O-acetyl-L-homoserine sulfhydrylase activities must first be introduced heterologously in enterobacteria such as E. coli, whereas these enzyme activities are naturally already present in corynebacteria such as C. glutamicum. The corresponding homologous or heterologous genes encoding the corresponding enzymes can each be enhanced by the measures described at the outset (such as increasing the copy number of both genes and/or the use of strong promoters).

[0027] In enterobacteria such as E. coli, the enhanced L-homoserine O-acetyltransferase and O-acetyl-L-homoserine sulfhydrylase activities may be introduced by transformation by means of suitable vectors comprising the gene sequences metX (e.g. SEQ ID No. 1) and metY (e.g. SEQ ID No. 3), which in each case is upstream of a strong promoter (for example Ptacl). An example of such a construct is the sequence according to sequence number 6 (SEQ ID No. 6).

[0028] The acetate liberated by the conversion of O-acetyl-L-homoserine to L-methionine, in the presence of methyl mercaptan (MC) and pyridoxal 5'-phosphate (PLP) and also the (heterologous) sulfhydrylase (MetY), is then used again for the synthesis of acetyl-CoA, with consumption of ATP, in the cytoplasm of E. coli (likewise Bacillus subtilis) by means of an acetate-inducible acetyl-CoA synthetase (Acs), which is activated particularly in the stationary phase or under anaerobic conditions by the regulator CsrA (S. Kumari et al., JOURNAL OF BACTERIOLOGY, Vol. 177, No. 10, May 1995, p. 2878-2886).

[0029] In contrast to E. coli, excess acetate in C. glutamicum is converted exclusively in an ATP-dependent reaction to acetyl phosphate, by means of an acetate kinase (AK), which finally reacts by means of a phosphotransacetylase (PTA) in the presence of CoA to give acetyl-CoA. The corresponding genes, ack and pta, are organized in C. glutamicum in an operon regulated by acetate at the transcriptional level (R. Gerstmeir et al. Journal of Biotechnology 104 (103) 99-122).

[0030] L-methionine is excreted out of the cell of E. coli by means of the YjeH exporter (Q. Liu et al., Appl Environ Microbiol 81 (2015) p. 7753-7766). Furthermore, the gene ygaZH in E. coli encodes a methionine exporter (WO2015/028675 A1). L-methionine is excreted out of the cell of C. glutamicum into the culture medium with the aid of the BrnFE exporter (C. Trötschel et al., JOURNAL OF BACTERIOLOGY, June 2005, p. 3786-3794).
Figure 1
Shows the pMW218 plasmid map.
Figure 2:
Shows the pMW218_Ptac-metX_Ptac-metY plasmid map.
Figure 3:
Shows the catalytic conversion of L-homoserine and acetyl-CoA by the cell homogenates of MG1655/pMW218 or MG1655/pMW218_Ptac-metX_Ptac-metY.
Figure 4:
Shows the catalytic conversion of sodium methyl mercaptide in the presence of O-acetylhomoserine and pyridoxal 5'-phosphate (PLP) by the O-acetylhomoserine sulfhydrylase enzyme (MetY). Comparison of the cell homogenates of MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY is shown with the respective total protein concentrations used.

Examples


1) Preparation of an enterobacterium heterologously expressing the genes for an L-homoserine O-acetyltransferase and a sulfhydrylase of a Corynebacteria species



[0031] On the basis of the genome sequence of Corynebacterium glutamicum (ATCC13032) NC_003450, the gene sequences metX (SEQ ID No. 1) and metY (SEQ ID No. 3), which encode the L-homoserine O-acetyltransferase having the amino acid sequence according to SEQ ID No. 2 and the O-acetyl-L-homoserine sulfhydrylase having the amino acid sequence according to SEQ ID No. 4 respectively, both with upstream promoter Ptacl (SEQ ID No. 5) (H.A. deBoer et al., Proc. Natl. Acad. Sci. USA, Vol. 80, 21-25, January 1983, Biochemistry) from Life Technologies Invitrogen GeneArt (Germany), were synthesized (SEQ ID No. 6).

[0032] In this SEQ ID No. 6, the Ptacl promoter is from base pair 407-447, the gene sequence of metX from 502-1638, the Ptacl promoter again from 1645-1685 and the gene sequence of metY from 1742-3055.

[0033] Subsequently, the cloning of this synthetic sequence was carried out via the restriction sites BssHII and BgII in the vector sequence pMW218 (Accession Number: AB005477) (Nippon Gene, Toyama, Japan) (Fig. 1). The plasmid pMW218_Ptac-metX_Ptac-metY is formed therefrom (Fig. 2). To analyze the pMW218_Ptac-metX_Ptac-metY plasmid, DNA sequencing was additionally carried out by Eurofins MWG Operon. The DNA sequences obtained were checked with respect to correctness using the Clone Manager software so that the nucleotide sequence SEQ ID No. 6 was confirmed.

[0034] The plasmids pMW218 and pMW218_Ptac-metX_Ptac-metY have been transformed in each case in the Escherichia coli K-12 strain MG1655 (DSM No. 18039). The transformants were subsequently cultured on LB medium agar plates with 50 µg/ml kanamycin such that the MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains could be generated. In each case a colony has been selected which was inoculated in each case into 10 ml of LB medium with 50 µg/ml kanamycin, and was cultured at 37°C, 200 rpm for 6 hours. Subsequently, 10 ml of medium A [25 g/l ammonium sulphate; 1 g/l magnesium sulphate heptahydrate; 2 g/l potassium dihydrogen phosphate; 0.03 g/l iron heptahydrate; 0.02 g/l manganese sulphate monohydrate; 20 g/l glucose monohydrate; 30 g/l calcium carbonate; 0.05 g/l kanamycin; 0.025 g/l pyridoxal 5'-phosphate (PLP); 0.0024 g/l isopropyl-β-D-thiogalactopyranoside (IPTG)] were inoculated with 200 µl of the growth cell culture and incubated at 37°C, 200 rpm for 16 h. These cell cultures were diluted with 10 ml of fresh medium A in a 100 ml flask to an OD of 2 and were further cultured under identical conditions until an OD of about 5 had been attained (circa 3-4 h). Subsequently, these cells, which are in the exponential growth phase and have homoserine O-acetyltransferase (MetX) and sulfhydrylase (MetY) activity, can be used for the biotransformation. Biotransformation is understood to mean a substance conversion, in which whole living cells, fixed cells or isolated free or carrier-linked enzymes or the combination of the above are used.

2) Detection of the enzymatic activities of L-homoserine O-acetyltransferase and acetyl-L-homoserine sulfhydrylase



[0035] 10 ml of LB medium with 50 µg/ml kanamycin have been inoculated in each case with a single colony of the MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains and have been cultured at 37°C, 200 rpm for 6 hours. Subsequently, 10 ml of medium A (see Example 1) were inoculated with 200 µl of the growth cell culture and incubated at 37°C, 200 rpm for 16 h. The cell cultures were then each harvested (8 ml normalized to an OD=1), the supernatants removed by centrifugation (20 min, 4000 rpm, 4°C) and the pelleted cells were washed twice with 800 µl of 0.1 M potassium phosphate buffer (pH 7.5) and taken up in 1 ml of buffer. The mechanical cell disruption was carried out in a FastPrep FP120 instrument (QBiogene, Heidelberg), wherein the cells were shaken three times for 20 s at 6.5 m/s in digestion vessels with 300 mg of glass beads (Ø 0.2-0.3 mm). The crude extract was then centrifuged at 12 000 rpm, 4°C, 20 min, in order to remove undigested cells and cell debris. The total amount of protein was determined using the Bio-Rad protein quantification assay (Bio-Rad, USA). The cell homogenate was then used for the enzymatic detection of the cytoplasmatic L-homoserine O-acetyltransferase and acetyl-L-homoserine sulfhydrylase activity.

2a) Detection of the cytoplasmatic activity of MetX (L-homoserine O-acetyltransferase)



[0036] The reaction, which the enzyme L-homoserine O-acetyltransferase (MetX) [EC2.3.1.31] catalyzes, is the conversion of L-homoserine and acetyl-CoA to O-acetyl-L-homoserine and CoA. With the aid of a DTNB solution (5,5'-dithiobis-2-nitrobenzoic acid, "Ellmans reagent", Sigma Aldrich, Germany) the progress of this reaction can be recorded by measurements of absorption at 412 nm, since DTNB forms a yellow substance with the SH group of CoA (S. Yamagata Journal of Bacteriology 169, No. 8 (1987) 3458-3463). The photometric MetX enzyme assay was conducted at 37°C, in which calibration was previously carried out using CoA concentrations between 0-200 µM. Each preparation was conducted in a 0.2 ml reaction mixture with 100 mM potassium phosphate buffer (pH 7.5), 0.65 mM DNTB [100 µl of a 1.3 mM DTNB stock], 0.13 mM acetyl-CoA [30 µl of a 0.886 mM acetyl-CoA stock, Sigma Aldrich, Germany], 10 mM L-homoserine [20 µl of a 100 mM L-homoserine stock, Sigma Aldrich, Germany] and the specified protein concentration of 0.012 mg/ml, or 0.024 mg/ml of the respective cell homogenate.

[0037] Since acetyl-CoA is used within the cell for various biosyntheses, diverse enzymes are present in the cytoplasm which catalyze the cleavage of acetyl-CoA to CoA, such that the difference between the cell homogenates with and without MetX needs to be considered.

[0038] It was observed as a result of the enzyme assay that the DNTB absorption increase of the cell homogenate of MG1655/pMW218_Ptac-metX_Ptac-metY was constantly above that of MG1655/pMW218 over the time course (Fig. 3). This confirms that MetX here is catalytically active as an additional enzyme. Comparison of the slopes of the initial linear regions of the recorded curves shows activities in the cell homogenate of MG1655/pMW218 of around 580 µmol/min per g of total protein and in MG1655/pMW218_Ptac-metX_Ptac-metY around 730 µmol/min per g of total protein. The difference therefore is the specific activity of the L-homoserine O-acetyltransferase (MetX) at around 150 units per g of total protein (1 unit = 1 µmol substrate conversion / min).

2b) Detection of the cytoplasmic activity of MetY (O-acetyl-L-homoserine sulfhydrylase)



[0039] The reaction, which the enzyme O-acetyl-L-homoserine sulfhydrylase (MetY) [EC 2.5.1.49] catalyzes, is the conversion of O-acetyl-L-homoserine with methanethiol (MC) in the presence of pyridoxal 5'-phosphate (PLP) to give L-methionine and acetate. As described in Example 2a, the progress of this reaction can be determined by means of DTNB absorption measurements at 412 nm, since DTNB reacts with the SH group of unreacted methyl mercaptan to give a yellow substance. For this purpose, the two strains MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY were prepared as cell homogenates as described above and the decrease or the conversion of the substrate sodium methyl mercaptide was measured in the subsequent enzyme assay.

[0040] Each preparation was conducted at 37°C in a 1 ml reaction mixture with 100 mM potassium phosphate buffer (pH 7.5), 2 mM sodium methyl mercaptide (NaMC) [10 µl of a 200 mM NaMC stock], 3 mM OAH HCI [30 µl of a 100 mM OAH HCI stock] and 0.01 mM PLP [10 µl of a 1 mM PLP stock] with the respective cell homogenate at a total protein concentration of 0.012 g/l; 0.024 g/l, or 0.048 g/l. Following the time-limited enzymatic reaction, the photometric measurement of the NaMC content by means of DTNB was conducted, wherein a calibration was previously carried out using MC concentrations between 0-200 µM. For this purpose, 180 µl of a DTNB solution (4 mg/ml) were added to each 20 µl of the enzymatic reaction mixture and subsequently measured at 412 nm.

[0041] The presence of the cell homogenate of the MG1655/pMW218_Ptac-metX_Ptac-metY strain leads to the decrease of the NaMC being catalyzed significantly faster than in the presence of the cell homogenate of MG1655/pMW218, due to the enzyme activity of MetY, depending on the total protein concentration (Figure 4). The decrease of NaMC likewise occurring, but more weakly, in the preparations with the cell homogenate of MG1655/pMW218 is independent of the amount of protein used. An identical decrease could also be observed in a preparation without cell homogenate and is due to the chemically dependent outgassing of the methyl mercaptan occurring from the solutions. From the difference in the slopes in the linear range, a specific MetY sulfhydrylase activity around 1500 units per g of total protein (1 unit = 1 µmol substrate conversion / min) can be calculated, which is on the same level as that from the MetX enzyme.

3) Detection of the cellular biotransformation of L-homoserine and sodium methyl mercaptide to give L-methionine



[0042] The MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains have been cultured as described in Example 1 and then, in the exponential phase of each preparation, have been adjusted to an OD600 of around 7.

[0043] The biotransformation was then carried out in 100 ml shaking flasks at 37°C, 200 rpm over a time period of 0, 2, 4 and 24 h. Each preparation was conducted in 10 ml of medium A with 6.5 g/l L-homoserine [500 µl of a 100 g/l homoserine stock] (Sigma Aldrich, Germany), 3 g/l NaMC [500 µl of a 6% NaMC stock] and 12 g/l KH2PO4 [600 µl of a 200 g/l KH2PO4 stock].

[0044] The conversion of L-homoserine with methyl mercaptan to give L-methionine was conducted using the MG1655/pMW218_Ptac-metX_Ptac-metY strain, whereas no L-methionine was synthesized using the MG1655/pMW218 strain (Table 1). The various yields based on the amounts of NaMC initially charged and amounts of L-homoserine consumed are based on an equal stoichiometry of both substrates present at the start but subsequent naturally occurring evaporation of the methyl mercaptan.
Table 1: Comparison of the biotransformations of 6.5 g/l synthetic L-homoserine and 3 g/l sodium methyl mercaptide using the MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains. The L-methionine titre obtained over the time course and the related yields are shown, based on the amount of NaMC pulsed at the start and the amount of L-homoserine (L-HS) consumed.
MG1655/pMW2180 h2 h4 h24 h
L-Met (g/l) <0.005 <0.005 <0.005 <0.005
         
MG1655/pMW218_Ptac-metX_Ptac-metY0 h2 h4 h24 h
L-Met (g/l) <0.005 0.810 0.980 1.500
L-Homoserine (g/l) 6.580 4.220 4.020 3.560
L-Met/initially charged NaMC (mol/mol) 0% 12.7% 15.3% 23.5%
L-Met/consumed L-HS (mol/mol) 0% 27.4% 30.6% 39.7%


[0045] Furthermore, a biotransformation using the MG1655/pMW218_Ptac-metX_Ptac-metY strain has been carried out, in which deuterated NaMC (D3CSNa) was used in place of the NaMC stock from Sigma Aldrich. This was prepared by introducing CD3SD (Sigma-Aldrich, 98 atom% D) into an equimolar amount of aqueous sodium hydroxide solution. (Alternatively, it can be prepared according to J. Voss et al., Phosphorous, Sulfur and Silicon and the Related Elements, 2012, 187, 382 from thiourea and CD3I.) Analysis of the solution after 24 h reaction by LC-MS showed a ratio of methionine to methionine-d-3 of 1:200. It could be detected, therefore, that the methionine formed in the biotransformation is formed exclusively by the incorporation of externally supplied methyl mercaptan.

4) Conversion of L-homoserine, produced by fermentation, to L-methionine via a biotransformation



[0046] On the basis of the biotransformation of synthetic L-homoserine to L-methionine conducted in Example 3a, the biotransformation of L-homoserine produced by fermentation has also been investigated. The concentration of the L-homoserine broth produced by fermentation was 10 g/l. The MG1655/pMW218_Ptac-metX_Ptac-metY strain has been cultured as in Example 1 and the biotransformation conducted in the exponential phase at an OD of 5 in the presence of 5 g/l L-homoserine produced by fermentation and as described in Example 3a for 2, 4 and 24 h. As shown in Table 2, after two hours' biotransformation around 7%, after four hours around 12% and after 24 hours around 45% of the substrates L-homoserine or NaMC were converted to L-methionine, which was reflected in a maximum titre of around 2.9 g/l L-methionine.
Table 2: L-methionine and the related yields formed by the biotransformation of 5 g/l L-homoserine produced by fermentation and 3 g/l sodium methyl mercaptide by the MG1655/pMW218_Ptac-metX_Ptac-metY strain.
Time (h)02424
L-Met (g/l) <0.005 0.45 0.79 2.87
L-Homoserine from fermentation (g/l) 5.26 4.74 4.24 2.15
L-Met/initially charged NaMC (mol/mol) 0% 7% 12% 45%
L-Met/consumed L-HS (mol/mol) 0% 69% 62% 74%

5) Cellular recycling of acetate during the biotransformation



[0047] To investigate the amounts of acetate formed in the biotransformation of L-homoserine and methyl mercaptan, preparations using the MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains in the presence of 5 g/l L-homoserine and 3 g/l sodium methyl mercaptide and 12 g/l KH2PO4 [600 µl of a 200 g/l KH2PO4 stock] were documented over four hours with respect to their acetate content.

[0048] The MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains have been prepared as described in Example 1, so that an exponential culture having a starting OD of around 3 was used for the respective 10 ml preparations in 100 ml flasks as described in Example 3b.

[0049] The acetate concentrations which are formed during the biotransformation of 5 g/l L-homoserine and 3 g/l sodium methyl mercaptide by the MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains are documented in Table 3. L-methionine is formed only in the preparation with the strain heterologously expressing the metX and metY genes, whereas in the preparation with the control strain MG1655/pMW218 no L-methionine was detectable.

[0050] Within the first four hours around 11 mM acetate are formed due to the experimental parameters in the control preparation, whereas in the biotransformation around 17 mM acetate and 7 Mm L-methionine are formed. The excess of acetate measured in the biotransformation preparation which gives rise to the difference is thus 6 mM. Due to the additional methionine synthesis with an equimolar production of acetate and methionine, which is not described for non-cellular systems (WO 2008/013432 A1), this value would be 7 mM. Therefore, a cellular recycling of the acetate formed in the L-methionine synthesis in the biotransformation could be detected. The additional acetate resulting from the biotransformation has therefore obviously been partly recycled by the acetyl-CoA synthetase (Acs) to acetyl-CoA.
Table 3: Formation and recycling of acetate in biotransformation preparations with the MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains in the presence of 5 g/l L-homoserine and 3 g/l sodium methyl mercaptide.
Time [h]MG1655/ pMW_Ptac-metX_Ptac-metYMG1655/ pMW_Ptac-metX_Ptac-metYMG1655/pMW218Excess acetate [mM]Amount of acetate expected (equimolar to L-Met) [mM]Recycled acetate [mM]
L-Met [mM]Acetate [mM]Acetate [mM]
0 <0.03 <0.08 <0.08 0.00 0.00 0.00
2 4.76 10.29 7.54 2.75 4.76 2.01
4 7.37 17.45 11.22 6.22 7.37 1.15

6) External addition of L-homoserine



[0051] The MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains have been prepared as described in Example 1, so that an exponential culture having a starting OD of around 3 was used for the respective 10 ml preparations in 100 ml flasks as described in Example 3b. To the respective preparations followed at the 0 h time point firstly no addition, the addition of 5 g/l L-homoserine, the addition of 3 g/l sodium methyl mercaptide with 12 g/l KH2PO4 [600 µl of a 200 g/l KH2PO4 stock] and the addition of 5 g/l L-homoserine with 3 g/l sodium methyl mercaptide and 12 g/l KH2PO4 [600 µl of a 200 g/l KH2PO4 stock].

[0052] The L-methionine and L-homoserine titres of the preparations were determined at the time points 0, 2, 4 and 6 h.
Table 4: L-methionine and L-homoserine titres after the external addition of 5 g/l L-homoserine (L-HS) to the biotransformation preparations using the MG1655/pMW218 and MG1655/pMW218_Ptac-metX_Ptac-metY strains in the presence and absence of 3 g/l sodium methyl mercaptide (Na-MC).
 MG1655/pMW_Ptac-metX_Ptac-metYMG1655/pMW218
Time [h]Addition L-HSNa-MC additionL-Methionine [g/l]L-Homoserine [g/l]L-Methionine [g/l]L-Homoserine [g/l]
0 - - <0.005 <0.005 <0.005 <0.005
2 - - <0.005 <0.005 <0.005 <0.005
4 - - <0.005 <0.005 <0.005 <0.005
6 - - <0.005 <0.005 <0.005 <0.005
24 - - <0.005 <0.005 <0.005 <0.005
             
0 5 g/l - <0.005 5.38 <0.005 5.34
2 - - <0.005 4.09 <0.005 5.08
4 - - <0.005 2.35 <0.005 4.77
6 - - <0.005 1.51 <0.005 4.46
24 - - <0.005 <0.005 <0.005 3.52
             
0 - 3 g/l <0.005 <0.005 <0.005 <0.005
2 - - <0.005 <0.005 <0.005 <0.005
4 - - 0.22 <0.005 <0.005 <0.005
6 - - 0.27 <0.005 <0.005 <0.005
24 - - 0.28 <0.005 <0.005 <0.005
             
0 5 g/l 3 g/l <0.005 5.01 <0.005 5.31
2     0.71 4.44 <0.005 5.09
4     1.10 4.11 <0.005 4.89
6     1.24 4.06 <0.005 4.65
24     1.46 3.56 <0.005 3.98

7) Biotransformation of L-homoserine and dimethyl disulfide (DMDS) or of L-homoserine and sodium methylmercaptide (NaMC) to give L-methionine



[0053] The MG1655/pMW218_Ptac-metX_Ptac-metY strain has been cultured as described in Example 1 and then, in the exponential phase of each preparation, has been adjusted to an OD600 of around 10.

[0054] The biotransformation was then carried out in 100 ml shaking flasks at 37°C, 200 rpm over a time period of 0, 24 and 48 h. Each preparation was conducted in 10 ml of medium A (see Example 1) with 5.0 g/l L-homoserine and 12 g/l KH2PO4 [600 µl of a 200 g/l KH2PO4 stock] and the quantities of the sulphur source (i.e. NaMC or DMDS) as provided in Table 5. The control did not contain any sulphur source (i.e. no NaMC and no DMDS).
Table 5: Comparison of the results of the biotransformation with NaMC, control (no sulphur source) and DMDS at different concentrations and reaction time periods
Sulphur source concentration, reaction timeL-Homoserine [g/l]L-Methionine [g/l]O-Acetyl-L-homoserine [g/l]
1.0 g/L NaMC, 24h 3.52 1.64 <0.005
Control, 0h 4.9 <0.005 <0.005
Control, 24h 1.48 <0.005 3.74
0.5 g/L DMDS, 24h 2.3 0.2 2.82
1 g/L DMDS, 24h 2.48 0.32 2.34
1 g/L DMDS, 48h 2.52 0.46 2.06
2 g/L DMDS, 24h 3.3 0.42 1.38

Sequences


SEQUENCE LISTING



[0055] 

<110> Evonik Degussa GmbH

<120> WHCB

<130> .

<160> 6

<170> PatentIn version 3.5

<210> 1
<211> 1137
<212> DNA
<213> Corynebacterium glutamicum

<220>
<221> CDS
<222> (1)..(1137)
<223> metX

<400> 1



<210> 2
<211> 377
<212> PRT
<213> Corynebacterium glutamicum

<400> 2



<210> 3
<211> 1314
<212> DNA
<213> Corynebacterium glutamicum

<220>
<221> CDS
<222> (1)..(1314)
<223> metY

<400> 3





<210> 4
<211> 437
<212> PRT
<213> Corynebacterium glutamicum

<400> 4





<210> 5
<211> 41
<212> DNA
<213> Hybrid promoter

<220>
<221> promoter
<222> (1)..(41)
<223> PtacI

<400> 5
gagctgttga caattaatca tcggctcgta taatgtgtgg a   41

<210> 6
<211> 3255
<212> DNA
<213> synthesized sequence

<400> 6








Claims

1. Method for producing L-methionine, wherein a microorganism having L-homoserine O-acetyltransferase activity and O-acetyl-L-homoserine sulfhydrylase activity is cultured in a culture medium comprising L-homoserine and a sulphur source, selected from the group consisting of methyl mercaptan, a methyl mercaptan salt and dimethyl disulfide, whereby L-methionine is accumulated in the culture medium.
 
2. Method according to Claim 1, wherein the microorganism is recombinant and the L-homoserine O-acetyltransferase activity and the O-acetyl-L-homoserine sulfhydrylase activity are enhanced.
 
3. Method according to Claim 2, wherein the L-homoserine O-acetyltransferase activity and the O-acetyl-L-homoserine sulfhydrylase activity are both enhanced by increased expression of a gene which encodes a protein having L-homoserine O-acetyltransferase activity or a protein having O-acetyl-L-homoserine sulfhydrylase activity.
 
4. Method according to Claim 3, wherein the increased gene expression is effected by increasing the copy number of the gene encoding the protein having L-homoserine O-acetyltransferase activity and/or the protein having O-acetyl-L-homoserine sulfhydrylase activity.
 
5. Method according to Claim 3 or 4, wherein the increased gene expression is effected by functional linkage of the gene encoding the protein having L-homoserine O-acetyltransferase activity and/or the protein having O-acetyl-L-homoserine sulfhydrylase activity to a strong promoter.
 
6. Method according to any of Claims 1 to 5, wherein the microorganism is selected from the group consisting of Enterobacteriaceae and Corynebacteriaceae.
 
7. Method according to any of Claims 1 to 6, wherein the L-homoserine O-acetyltransferase activity is the enzyme MetX, which originates from Corynebacterium glutamicum.
 
8. Method according to any of Claims 1 to 7, wherein O-acetyl-L-homoserine sulfhydrylase activity is the enzyme MetY, which originates from Corynebacterium glutamicum.
 


Ansprüche

1. Verfahren zur Herstellung von L-Methionin, bei welchem ein Mikroorganismus, der L-Homoserin-O-Acetyltransferase-Aktivitat und O-Acetyl-L-Homoserin-Sulfhydrylase-Aktivitat aufweist, in einem Kulturmedium umfassend L-Homoserin und eine Schwefelquelle, ausgewählt aus der Gruppe bestehend aus Methylmercaptan, einem Methylmercaptan-Salz und Dimethyldisulfid, kultiviert wird, wobei sich L-Methionin im Kulturmedium anreichert.
 
2. Verfahren nach Anspruch 1, wobei der Mikroorganismus rekombinant ist und die L-Homoserin-O-Acetyltransferase-Aktivitat und die 0-Acetyl-L-Homoserin-Sulfhydrylase-Aktivitat verstärkt sind.
 
3. Verfahren nach Anspruch 2, wobei die L-Homoserin-O-Acetyltransferase-Aktivitat und die 0-Acetyl-L-Homoserin-Sulfhydrylase-Aktivitat jeweils durch eine erhohte Expression eines Gens, welches fur ein Protein mit L-Homoserin-O-Acetyltransferase-Aktivitat beziehungsweise fur ein Protein mit 0-Acetyl-L-Homoserin-Sulfhydrylase-Aktivitat kodiert, verstarkt werden.
 
4. Verfahren nach Anspruch 3, wobei die erhohte Genexpression durch Erhohung der Kopienzahl des fur das Protein mit L-Homoserin-O-Acetyltransferase-Aktivitat und/oder fur das Protein mit 0-Acetyl-L-Homoserin-Sulfhydrylase-Aktivitat kodierenden Gens bewirkt wird.
 
5. Verfahren nach Anspruch 3 oder 4, wobei die erhohte Genexpression durch funktionelle VerknOpfung des für das Protein mit L-Homoserin-O-Acetyltransferase-Aktivitat und/oder für das Protein mit 0-Acetyl-L-Homoserin-Sulfhydrylase-Aktivitat kodierenden Gens mit einem starken Promotor bewirkt wird.
 
6. Verfahren nach einem der Anspruche 1 bis 5, wobei der Mikroorganismus ausgewahlt ist aus der Gruppe bestehend aus Enterobacteriaceae und Corynebacteriaceae.
 
7. Verfahren nach einem der Anspruche 1 bis 6, wobei die L-Homoserin-O-Acetyltransferase-Aktivitat das Enzym MetX ist, welches aus Corynebacterium glutamicum stammt.
 
8. Verfahren nach einem der Anspruche 1 bis 7, wobei 0-Acetyl-L-Homoserin-Sulfhydrylase- Aktivitat das Enzym MetY ist, welches aus Corynebacterium glutamicum stammt.
 


Revendications

1. Procédé de production de L-méthionine, un microorganisme possédant une activité L-homosérine 0-acétyltransférase et une activité O-acétyl-L-homosérine sulfhydrylase étant cultivé dans un milieu de culture comprenant de la L-homosérine et une source de soufre, choisie dans le groupe constitué par le méthylmercaptan, un sel de méthylmercaptan et le disulfure de diméthyle, la L-méthionine s'accumulant dans le milieu de culture.
 
2. Procédé selon la revendication 1, le microorganisme étant recombinant et l'activité L-homosérine 0-acétyltransférase et l'activité O-acétyl-L-homosérine sulfhydrylase étant augmentées.
 
3. Procédé selon la revendication 2, l'activité L-homosérine O-acétyltransférase et l'activité O-acétyl-L-homosérine sulfhydrylase étant toutes deux augmentées par l'amplification de l'expression d'un gène qui code pour une protéine ayant une activité L-homosérine O-acétyltransférase ou pour une protéine ayant une activité O-acétyl-L-homosérine sulfhydrylase.
 
4. Procédé selon la revendication 3, l'amplification de l'expression génique étant réalisée par amplification du nombre de copies du gène codant pour la protéine ayant une activité L-homosérine O-acétyltransférase et/ou pour la protéine ayant une activité O-acétyl-L-homosérine sulfhydrylase.
 
5. Procédé selon la revendication 3 ou 4, l'amplification de l'expression génique étant réalisée par liaison fonctionnelle du gène codant pour la protéine ayant une activité L-homosérine O-acétyltransférase et/ou pour la protéine ayant une activité O-acétyl-L-homosérine sulfhydrylase à un promoteur fort.
 
6. Procédé selon l'une quelconque des revendications 1 à 5, le microorganisme étant choisi dans le groupe constitué par Enterobacteriaceae et Corynebacteriaceae.
 
7. Procédé selon l'une quelconque des revendications 1 à 6, l'activité L-homosérine O-acétyltransférase venant de l'enzyme MetX, qui provient de Corynebacterium glutamicum.
 
8. Procédé selon l'une quelconque des revendications 1 à 7, l'activité O-acétyl-L-homosérine sulfhydrylase venant de l'enzyme MetY, qui provient de Corynebacterium glutamicum.
 




Drawing











Cited references

REFERENCES CITED IN THE DESCRIPTION



This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description




Non-patent literature cited in the description