WO2018100097A1 - Alcool acétyl-transférases pour la production d'acétate d'éthyle - Google Patents

Alcool acétyl-transférases pour la production d'acétate d'éthyle Download PDF

Info

Publication number
WO2018100097A1
WO2018100097A1 PCT/EP2017/081048 EP2017081048W WO2018100097A1 WO 2018100097 A1 WO2018100097 A1 WO 2018100097A1 EP 2017081048 W EP2017081048 W EP 2017081048W WO 2018100097 A1 WO2018100097 A1 WO 2018100097A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
amino acid
identity
nucleotide sequence
acid sequence
Prior art date
Application number
PCT/EP2017/081048
Other languages
English (en)
Inventor
Aleksander Johannes KRUIS
Mark LEVISSON
Astrid Elisabeth MARS
Servatius Wilhelmus Maria KENGEN
John Van Der Oost
Johan Pieter Marinus Sanders
Ruud Alexander Weusthuis
Original Assignee
Akzo Nobel Chemicals International B.V.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Akzo Nobel Chemicals International B.V. filed Critical Akzo Nobel Chemicals International B.V.
Publication of WO2018100097A1 publication Critical patent/WO2018100097A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1025Acyltransferases (2.3)
    • C12N9/1029Acyltransferases (2.3) transferring groups other than amino-acyl groups (2.3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/62Carboxylic acid esters

Definitions

  • the invention relates to the use of a polypeptide having alcohol acetyl transferase activity (hereinafter referred to as A AT), novel AAT for the microbial production of ethyl acetate from alcohol and acetyl coenzyme A, and to a method for the production of ethyl acetate.
  • a AT alcohol acetyl transferase activity
  • novel AAT for the microbial production of ethyl acetate from alcohol and acetyl coenzyme A
  • the present invention further relates to a recombinant expression vector or plasmid or host cell comprising said nucleotide sequence. More particularly, the polypeptide has alcohol acetyl transferase activity, and catalyzes the transfer of the acetyl group of acetyl coenzyme A to an alcohol (EC2.3.1.84) and produces coenzyme A and the corresponding ester.
  • Ethyl acetate is an environmentally friendly solvent with many industrial applications.
  • the production of ethyl acetate currently proceeds by energy-intensive petrochemical processes which are based on natural gas and crude oil without exception.
  • the following methods play a larger role in industrial-scale production (Lin et al. 1998: Colley et al. 2004; Arpe 2007):
  • Fischer esterification reaction of ethanol with acetic acid in the presence of strong acids as a catalyst, in part combined with removal of water for shifting the equilibrium toward the ester
  • Tischtschenko reaction addition of two equivalents acetaldehyde by disproportionation under the action of alkoxides, especially aluminum triethoxide, as a catalyst
  • ethanol is dehydrogenated to acetaldehyde, the aldehyde is then added to ethanol to form a hemiacetal, which in turn is dehydrogenated to generate ethyl acetate (Inui et al. 2002),
  • Avada process developed by BP Chemicals the abbreviation Avada stands for "Advanced acetates by direct addition” and means synthesis of ethyl acetate by catalytic addition of ethylene and acetic acid in the gas phase,
  • Ethanol as a precursor for chemical synthesis of ethyl acetate could be produced from sugar by fermentation (Silveira et al. 2005; Aziz et al. 2009; Guimaraes et al. 2010; Rodrussamee et al. 201 1 ); however, the direct microbial production of ethyl esters such as ethyl acetate would provide more advantages. Microbial synthesis of ethyl acetate could become an interesting alternative. Although the ability of yeasts to produce larger amounts of this ester has been known for a long time, these native microorganisms are not able to produce acetate esters in yields that are suitable for industrial bulk production.
  • ethyl acetate has been associated with three enzymatic reactions: Esterase reaction, wherein ethanol and acetate forms ethyl acetate with formation of water. Hemiacetal dehydrogenase reaction (HADH), wherein ethanol and acetaldehyde form hemiacetal, which is formed into ethyl acetate and
  • AAT Alcohol acetyl transferase
  • Eht1 and Eeb1 produce medium-chain ethyl esters in S. cerevisiae. They do not resemble Atf 1 and Atf2 on a protein level and contain an alpha/beta hydrolase fold.
  • the reaction typically associated with alpha/beta hydrolases is hydrolysis, but Eht1 and Eeb1 show AAT, thioesterase and esterase activities.
  • Eht1 and Eeb1 are not known to produce ethyl acetate, but since no combined gene
  • An object of this invention is to provide a polypeptide, as well as its gene sequence, with alcohol acetyl transferase activity that can be used for the microbial production of ethyl acetate.
  • the inventors have found a group of homologous polypeptides which have this alcohol acetyl transferase activity.
  • the genes encoding these polypeptides, when expressed in a suitable host, were able to produce ethyl acetate in a yield suitable for use on an industrial scale.
  • This group of polypeptides was found to represent enzymes that have ethyl acetate-producing alcohol acetyl transferase activity and whose three-dimensional structure contains an alpha-beta hydrolase fold and an active site comprising a serine, histidine, and optionally aspartate forming a dyad of serine- histidine or a triad of serine-aspartic acid-histidine.
  • the histidine of the serine-histidine dyad or serine-aspartic acid-histidine triad in the polypeptide according to the invention is present in a polypeptide fragment that has a glutamic acid-arginine-proline (ERP) fragment, a glutamic acid-asparagine-proline (ENP) fragment, or a glutamic-acid-methionine-proline (EMP) fragment as the 5 th , 6 th and 7 th amino acid from the histidine on the C-terminal side of the polypeptide.
  • EMP glutamic-acid-methionine-proline
  • amino acid sequence of the polypeptide used in the process according to the disclosure comprises an amino acid sequence that has: - an amino acid sequence according to SEQ ID NO: 1 , or an amino acid sequence that has at least 70% identity with the sequence as shown in SEQ ID NO: 1 and/or
  • nucleotide sequence encoding for the polypeptide used in the process according to the disclosure comprises a nucleotide sequence according to SEQ ID NO:2, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 2 and/or
  • nucleotide sequence according to SEQ ID NO:4, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 4 and/or
  • nucleotide sequence according to SEQ ID NO:6, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 6 and/or
  • nucleotide sequence according to SEQ ID NO:8, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 8 and/or
  • nucleotide sequence according to SEQ ID NO:10, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 10 and/or
  • nucleotide sequence according to SEQ ID NO:12, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 12 and/or
  • nucleotide sequence according to SEQ ID NO:14, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 14 and/or,
  • nucleotide sequence according to SEQ ID NO:16, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 16 and/or
  • nucleotide sequence according to SEQ ID NO:18, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 18 and/or
  • nucleotide sequence according to SEQ ID NO:20, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 20.
  • the invention is further directed to the process using a recombinant vector, a plasmid or a host cell comprising the nucleotide sequence according to the invention.
  • Said host cell may be selected from bacteria, yeasts and filamentous fungi.
  • Preferred bacteria are selected from Escherichia species and Bacillus species.
  • a preferred Escherichia species is E. coli and a preferred Bacillus species is Bacillus subtilis.
  • Other suitable bacteria are the ones used in industrial production such as Corynebacterium glutamicum.
  • thermophilic species since they allow production at high temperatures.
  • examples thereof are Geobacillus thermoglucosidasius, Caldiceliulosiruptor bescii, Clostridium thermocellum, Thermoanaerobacterium aotearoense, Thermoanaerobacterium saccharolyticum, Thermoanaerobacter tengcongensis, Thermoanaerobacter ethanolicus, Thermoanaerobacter mathrani, Thermococcus kodakarensis, Pyrococcus furiosus.
  • Preferred yeasts are the ones from which the polypeptides according to the invention originate such as Wickerhamomyces anomalus, Wickerhamomyces ciferrii, Kluyveromyces marxianus, Kluyveromyces lactis, Cyberlindnera jadinii, Hanseniaspora uvarum, Eremothecium cymbalariae, but also yeasts that are often used in the industry as production yeasts, such as Saccharomyces cerevisiae species, Pichia species such as P. pastoris and Schizosaccharomyces species.
  • Preferred filamentous fungi are selected from Aspergillus species, Trichoderma species or Penicillium species; Preferred Aspergillus species are A. niger, A. oryzae, and A. nidulans. Also Monascus ruber is suitable.
  • the invention is directed to a process for the production of ethyl acetate using the polypeptide according to the invention to convert acetyl coenzyme A and ethanol into ethyl acetate.
  • the process may be conducted at a temperature of between 10 and 130 °C.
  • the polypeptides used in the process are produced in a host cell.
  • the ethanol and the acetyl coenzyme A may also be produced in a host cell.
  • the ethyl acetate formed may be isolated from the host cell and optionally purified.
  • the invention is further directed to the use of the hosts cells according to the invention in the preparation of ethyl acetate.
  • a sugar compound may be used as a growth substrate for the host cell.
  • SEQ ID NO:1 sets out the amino acid sequence of AAT1 obtained from Wickerhamomyces anomalus DSM 6766.
  • SEQ ID NO:2 sets out the nucleotide sequence encoding for AAT1.
  • SEQ ID NO:3 sets out the amino acid sequence of AAT 2 obtained from Wickerhamomyces ciferrii CBS 11 1.
  • SEQ ID NO:4 sets out the nucleotide sequence encoding for AAT2.
  • SEQ ID NO:5 sets out the amino acid sequence of AAT 3 obtained from Kluyveromyces marxianus
  • SEQ ID NO:6 sets out the nucleotide sequence encoding for AAT3.
  • SEQ ID NO:7 sets out the amino acid sequence of AAT4 obtained from Kluyveromyces lactis CBS 2359.
  • SEQ ID NO:8 sets out the nucleotide sequence encoding for AAT4.
  • SEQ ID NO:9 sets out the amino acid sequence of AAT 5 obtained from Cyberlindnera jadinii DSM 2361.
  • SEQ ID NO:10 sets out the nucleotide sequence encoding for AAT5.
  • SEQ ID NO:1 1 sets out the amino acid sequence of AAT 6 obtained from Cyberlindnera fabianii CBS 5640).
  • SEQ ID NO:12 sets out the nucleotide sequence encoding for AAT6.
  • SEQ ID NO:13 sets out the amino acid sequence of AAT7 obtained from Hanseniaspora u varum CECT 1 1 105.
  • SEQ ID NO:14 sets out the nucleotide sequence encoding for AAT7.
  • SEQ ID NO:15 sets out the amino acid sequence of AAT 8 obtained from Hanseniaspora u varum CECT 1 1 105.
  • SEQ ID NO:16 sets out the nucleotide sequence encoding for AAT8.
  • SEQ ID NO:17 sets out the amino acid sequence of AAT 9 obtained from Eremothecum cymbalariae.
  • SEQ ID NO:18 sets out the nucleotide sequence encoding for AAT 9.
  • SEQ ID NO:19 sets out the amino acid sequence of AAT 10 obtained from Saccharomyces cerevisiae NCYC 2629.
  • SEQ ID NO:20 sets out the nucleotide sequence encoding for AAT10. DETAILED DESCRIPTION OF THE INVENTION
  • a preferred polypeptide that has enzyme activity according to the invention was encoded by a gene isolated from the genome of W. anomalus, hereinafter referred to as AAT 1 .
  • the amino sequence of AAT 1 is given under SEQ ID No:1
  • the nucleotide sequence encoding for AAT 1 is given in SEQ ID NO: 2.
  • homologs of AAT 1 Upon investigating homologs of AAT 1 according to the invention it was found that these homologs contain a highly conserved nucleophilic elbow (GYSLG) at Ser 121 . Moreover, Asp 145, Asp 178 and His 295 were also highly conserved. It was found that homologs of AAT 1 had the same highly conserved serine, aspartic acid and histidine. Upon visualization with a three-dimensional model of AAT 1 , Ser 121 , Asp 145 and His 295 were shown to be in the correct spatial proximity to form a catalytic Ser-Asp-His triad that occurs in alpha/beta hydrolases. Some homologs however did not have the aspartic acid at a corresponding site.
  • GYSLG nucleophilic elbow
  • polypeptides were found to be enzymes that have alcohol acetyl transferase activity and n alpha-beta hydrolase fold and an active site comprising a serine and histidine dyad or a serine, aspartic acid, histidine triad in their three-dimensional structure.
  • This triad and alpha/beta hydrolase fold is not present in the known aftl and aft2 enzymes of S. cerevisiae. It was found that polypeptides with ethyl acetate-producing AAT activity and the structural features as described above had a good yield in ethyl acetate production when expressed in a proper host cell.
  • polypeptides to be used according to the invention are genes isolated from Kluyveromyces marxianus, Kluyveromyces lactis, Wickerhamomyces ciferrii, Cyberlindnera jadinii, Hanseniaspora uvarum, Saccharomyces cerevisiae and Eremothecium cymbalariae. These genes showed roughly 50% identity to AAT 1 , but some of them showed even less identity with AAT 1 . Upon expression in a host, these genes also have ethyl acetate producing AAT activity, similar to AAT 1. These new enzymes are referred to as AAT 2 to AAT 10.
  • amino sequences of these AAT 2-10 are given under SEQ ID No:3, SEQ ID No:5, SEQ ID No:7, SEQ ID No:9, SEQ ID No:1 1 , SEQ ID No:13, SEQ ID No:15, SEQ ID No:17 and SEQ ID No: 19, respectively.
  • the nucleotide sequences encoding for AAT 2-10 are given in SEQ ID NO: 4, SEQ ID No: 6, SEQ ID NO: 8, SEQ ID No: 10, SEQ ID NO: 12, SEQ ID No: 14, SEQ ID NO: 16, SEQ ID No: 18, and SEQ ID NO: 20, respectively.
  • polypeptides according to the invention which have: - an amino acid sequence according to SEQ ID NO: 1 , or an amino acid sequence that has at least 70% identity with the sequence as shown in SEQ ID NO: 1 and/or
  • polypeptides have the structural features as described above.
  • sequence identity in order to determine the degree of sequence identity shared by two amino acid sequences or by two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). Such alignment may be carried out over the full lengths of the sequences being compared. Alternatively, the alignment may be carried out over a shorter comparison length, for example over about 20, about 50, about 100 or more nucleic acids/bases or amino acids.
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the two sequences being compared are of the same or substantially the same length.
  • the skilled person will be aware of the fact that several different computer programs are available to determine the homology between two sequences. For instance, a comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. In a preferred embodiment, the percentage identity between two amino acid sequences is determined using the Needleman and Wunsch (J. Mol. Biol. (48): 444-453 (1970)) algorithm which has been incorporated into the GAP program in the Accel rys GCG software package (available at http://www.accelrys.com/products/qcq/), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1 , 2, 3, 4, 5, or 6. The skilled person will appreciate that all these different parameters will yield slightly different results but that the overall percentage identity of two sequences is not significantly altered when using different algorithms.
  • the polypeptide comprises a substantially homologous amino acid sequence that has at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity with any of the amino acid sequences shown in SEQ ID NO: 1 and/or SEQ ID NO:3, and/or SEQ ID NO 5, and/or SEQ ID NO: 7 and/or SEQ ID NO:9, and/or SEQ ID NO 1 1 , and/or SEQ ID NO: 13 and/or SEQ ID NO:15, and/or SEQ ID NO 17, and/or SEQ ID NO 19.
  • the enzyme may be a polypeptide derived from amino acid SEQ ID NO: 1 to 19 by substitution, deletion or addition of one or several amino acid residues in any of the amino acid sequences of SEQ ID NO: 1 and/or SEQ ID NO:3, and/or SEQ ID NO 5, and/or SEQ ID NO: 7 and/or SEQ ID NO:9, and/or SEQ ID NO 1 1 , and/or SEQ ID NO: 13 and/or SEQ ID NO:15, and/or SEQ ID NO 17, and or SEQ ID NO 19 and having the same activity as any of the amino acid residue sequences of SEQ ID NO: 1 -19.
  • the nucleotide sequence encoding for the polypeptide used according to the invention comprises: a nucleotide sequence according to SEQ ID NO:2, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 2 and/or
  • nucleotide sequence according to SEQ ID NO:4, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 4 and/or
  • nucleotide sequence according to SEQ ID NO:6, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 6 and/or
  • nucleotide sequence according to SEQ ID NO:8, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 8 and/or
  • nucleotide sequence according to SEQ ID NO:10, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 10 and/or
  • nucleotide sequence according to SEQ ID NO:12, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 12 and/or
  • nucleotide sequence according to SEQ ID NO:14, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 14 and/or
  • nucleotide sequence according to SEQ ID NO:16, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 16 and/or
  • nucleotide sequence according to SEQ ID NO:18, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 18 and/or
  • nucleotide sequence according to SEQ ID NO:20, or a nucleotide sequence having at least 70% identity with SEQ ID NO: 20.
  • the nucleic acid molecule comprises a nucleotide sequence encoding a protein, comprising a substantially homologous nucleotide sequence of at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more homologous to the nucleotide sequences shown in nucleotide SEQ ID NO: 2 and/or SEQ ID NO: 4 and/or SEQ ID NO: 6, SEQ ID NO: 8 and/or SEQ ID NO: 10 and/or SEQ ID NO: 12, SEQ ID NO: 14 and/or SEQ ID NO: 16 and/or SEQ ID NO: 18, SEQ ID NO: 20.
  • the present invention provides a gene which hybridizes selectively under stringent conditions to all or part of the DNA as shown in any of nucleotide SEQ ID NO: 2-20 to all and/or part of a DNA complementary to the sequence as shown in any of nucleotide SEQ ID NO: 2-20 and which encodes a protein having the activity of alcohol acetyl transferase.
  • hybridizes selectively As used herein “selectively hybridizing”, “hybridizes selectively” and similar terms are intended to describe conditions for hybridization and washing under which nucleotide sequences of at least 66%, at least 70%, at least 75%, at least 80%, more preferably at least 85%, even more preferably at least 90%, preferably at least 95%, more preferably at least 98% or more preferably at least 99% homologous to each other typically remain hybridized to each other.
  • hybridizing sequences may share at least 45%, at least 50%, at least 55%, at least 60%, at least 65, at least 70%, at least 75%, at least 80%, more preferably at least 85%, even more preferably at least 90%, more preferably at least 95%, more preferably at least 98% or more preferably at least 99% sequence identity.
  • a preferred, non-limiting example of such stringent hybridization conditions is hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45 q C, followed by one or more washes in 1 X SSC, 0.1 % SDS at about 50 °C, preferably at about 55 °C, preferably at about 60 ⁇ and even more preferably at about 65 °C.
  • Highly stringent conditions include, for example, hybridization at about 68 ⁇ € in 5x SSC/5x Denhardt's solution / 1 .0% SDS and washing in 0.2x SSC/0.1 % SDS at room temperature. Alternatively, washing may be performed at 42 q C. The skilled artisan will know which conditions to apply for stringent and highly stringent hybridization.
  • a polynucleotide which hybridizes only to a poly A sequence such as the 3' terminal poly(A) tract of mRNAs), or to a complementary stretch of T (or U) residues, would not be included in a polynucleotide of the invention used to specifically hybridize to a portion of a nucleic acid of the invention, since such a polynucleotide would hybridize to any nucleic acid molecule containing a poly (A) stretch or the complement thereof (e.g., practically any double-stranded cDNA clone).
  • Homologous gene sequences can be isolated, for example, by performing PGR using two degenerate oligonucleotide primer pools designed on the basis of nucleotide sequences as taught herein.
  • the template for the reaction can be total chromosomal DNA from the strain known or suspected to express a polynucleotide according to the invention.
  • the PGR product can be subcloned and sequenced to ensure that the amplified sequences represent the sequences of a new oxidoreductase nucleic acid sequence, or a functional equivalent thereof.
  • the template for the reaction can be cDNA obtained by reverse transcription of mRNA prepared from strains known or suspected to express a polynucleotide according to the invention.
  • the PGR product can be subcloned and sequenced to ensure that the amplified sequences represent the sequences of a new oxidoreductase nucleic acid sequence, or a functional equivalent thereof.
  • the PGR fragment can then be used to isolate a full-length cDNA clone by a variety of known methods.
  • the amplified fragment can be labeled and used to screen a bacteriophage or cosmic cDNA library.
  • the labeled fragment can be used to screen a genomic library.
  • RNA can be isolated, following standard procedures, from an appropriate cellular or tissue source.
  • a reverse transcription reaction can be performed on the RNA using an oligonucleotide primer specific for the most 5' end of the amplified fragment for the priming of first strand synthesis.
  • the resulting RNA/DNA hybrid can then be "tailed" (e.g., with guanines) using a standard terminal transferase reaction, the hybrid can be digested with RNase H, and second strand synthesis can then be primed (e.g., with a poly-C primer).
  • cDNA sequences upstream of the amplified fragment can easily be isolated.
  • vectors including cloning and expression vectors, comprising a polynucleotide of the invention encoding alcohol acetyl transferase protein or a functional equivalent thereof and methods of growing, transforming or transfecting such vectors in a suitable host cell, for example under conditions in which expression of a polypeptide of the invention occurs.
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • nucleotide sequence present in a host organism, microorganism or cell refers to a nucleotide sequence which is not naturally present in said organism, microorganism or cell. This includes nucleotide sequences which are foreign to said organism, microorganism or cell and nucleotide sequences which are introduced at a position other than their natural position in the genome and endogenous gene sequences which have been modified.
  • heterologous is used herein with reference to a nucleotide sequence present in a host organism, microorganism or cell and refers to a nucleotide sequence that has been derived from a different organism.
  • polynucleotides of the invention can be incorporated into a recombinant replicable vector, for example a cloning or expression vector.
  • the vector may be used to replicate the nucleic acid in a compatible host cell.
  • the invention provides a method of making polynucleotides of the invention by introducing a polynucleotide of the invention into a replicable vector, introducing the vector into a compatible host cell, and growing the host cell under conditions which bring about replication of the vector.
  • the vector may be recovered from the host cell. Suitable host cells are described below.
  • the vector into which the expression cassette or polynucleotide of the invention is inserted may be any vector which may conveniently be subjected to recombinant DNA procedures, and the choice of the vector will often depend on the host cell into which it is to be introduced.
  • a vector according to the invention may be an autonomously replicating vector, i.e. a vector which exists as an extra-chromosomal entity, the replication of which is independent of chromosomal replication, e. g. a plasmid.
  • the vector may be one which, when introduced into a host cell, is integrated into the host cell genome and replicated together with the chromosome(s) into which it has been integrated.
  • vector refers to a circular double-stranded DNA loop into which additional DNA segments can be ligated.
  • plasmid refers to a circular double-stranded DNA loop into which additional DNA segments can be ligated.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • Other vectors e.g., non-episomal mammalian vectors
  • certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "expression vectors”.
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and “vector” can be used interchangeably herein as the plasmid is the most commonly used form of vector.
  • the invention is intended to include other forms of expression vectors, such as cosmid, viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses) and phage vectors, which serve equivalent functions.
  • Vectors according to the invention may be used in vitro, for example for the production of RNA or used to transfect or transform a host cell.
  • a vector of the invention may comprise two or more, for example three, four or five, polynucleotides of the invention, for example for overexpression.
  • the recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the
  • recombinant expression vector includes one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which are operably linked to the nucleic acid sequence to be expressed.
  • operably linked is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell), i.e. the term “operably linked” refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner.
  • a regulatory sequence such as a promoter, enhancer or other expression regulation signal "operably linked" to a coding sequence is positioned in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences or the sequences are arranged so that they function in concert for their intended purpose, for example transcription initiates at a promoter and proceeds through the DNA sequence encoding the polypeptide according to the invention.
  • the present invention also provides a recombinant vector or a plasmid containing the nucleotide sequence encoding for the polypeptide for use according to the invention.
  • Said plasmid can be introduced into proper host cells.
  • Said host cell may contain a homologous, or a heterologous gene, which is capable of generating an amino acid sequence that has at least 70% identity with the sequence shown in any of the amino acid SEQ ID NO: 1 -19.
  • this gene may be incorporated into the host cell in such a way as to increase the ethyl acetate yield compared to the ethyl acetate yield that can be obtained via the gene in its natural position in the genome and with endogenous gene sequences, i.e. overexpression.
  • endogenous gene sequences i.e. overexpression.
  • the present invention further provides proper host cells, which comprise a nucleotide sequence exhibiting at least 70% identity with that shown in any of the nucleotide SEQ ID NO: 2-20.
  • Suitable host organisms are selected from bacteria, yeasts, or filamentous fungi.
  • Preferred bacteria are selected from Escherichia species and Bacillus species.
  • a preferred Escherichia species is preferably E. coli and a preferred Bacillus species is Bacillus subtilis.
  • Other suitable bacteria are the ones used in industrial production such as Corynebacterium glutamicum. Species that are especially preferred are the thermophilic species since they allow production at high temperatures. Examples thereof are Geobacillus thermoglucosidasius. Caldicellulosiruptor bescii, Clostridium thermocellum, Thermoanaerobacterium aotearoense,
  • yeasts are the ones from which the polypeptides according to the invention originate, such as Wickerhamomyces anomalus, Wickerhamomyces ciferrii, Kluyveromyces marxianus, Kluyveromyces lactis, Cyberlindnera jadinii, Hanseniaspora uvarum, Eremothecium cymbalariae, but also yeasts that are often used in the industry as production yeasts such as Saccharomyces cerevisiae species, Pichia species such as P. pastoris and Schizosaccharomyces species.
  • Preferred filamentous fungi are selected from Aspergillus species, Trichoderma species or Penicillium species; preferred Aspergillus species are A. niger, A. oryzae, and A. nidulans. Also Monascus ruber is suitable. Appropriate culture media and conditions for the above-described host cells are known in the art.
  • the present invention is also directed to the use of host cells comprising the nucleotide sequence of this invention in the preparation of ethyl acetate ester by reacting ethanol and acetyl coenzyme A in the presence of the alcohol acetyl transferase to form ethyl acetate.
  • the acetyl coenzyme A may be produced by the host cell.
  • the ethanol can either be added to the reaction or the conditions are arranged such that the ethanol is also produced in the host cell.
  • suitable host cells were transformed with said DNA by conventional methods in the field, and the recombinant enzyme produced by the recombinant cells after transformation converts ethanol and acetyl coenzyme A to ethyl acetate in the host cell.
  • the invention is directed to a process for the production of acetate ester wherein the polypeptide according to the invention converts ethanol and acetyl coenzyme A into ethyl acetate.
  • the polypeptide to be used according to the invention may be produced in a host cell.
  • the formed polypeptide may be used after isolation from the host cell, or the process may be a microbial process, taking place in the host cell.
  • the process may also be arranged so that the ethanol needed to form ethyl acetate is formed by fermentation of sugar compounds either separately from the conversion to ethyl acetate or simultaneously.
  • the process is conducted as a full microbial process wherein both ethanol and acetyl coenzyme A are formed within the host cell and converted into ethyl acetate.
  • the process according to the invention may be conducted at a temperature of between 10 and 130 °C. This has clear advantages over any known chemical process for the production of ethyl acetate, where much higher temperatures, higher amounts of energy and increased pressures are needed.
  • Thermophilic hosts allow the process to be conducted at relatively high temperature such as between 45 and 130 °C, preferably between 50 and 100 °C. It goes without saying that the polypeptide to be incorporated into a thermophilic host should be able to undergo the growth and production temperatures used.
  • Preferred sugar compounds to be used as growth substrates are selected from compounds comprising both hexose and pentose sugars.
  • the sugar compound may be a hexose sugar such as glucose, fructose, galactose, glycan or other polymers of glucose, hexose oligomers such as sucrose, lactose, maltose, maltotriose and isomaltotriose, panose, and fructose oligomers.
  • the micro-organism is modified to have the ability to ferment pentose sugars, and the medium includes a pentose sugar such as xylose, xylan or other oligomer of xylose.
  • the organisms are cultivated on combinations of hexose and pentose sugars.
  • the sugars can be hydrolysates of a hemiceilulose or cellulose-containing biomass.
  • the micro-organism is modified to ensure degradation of the biomass to monomers (e.g. expression of cellulase genes).
  • the substrate comprises a sugar oligomer or polymer such as cellulose, hemiceilulose or pectin.
  • enzymes can be added to the fermentation medium to ensure degradation of the substrate into fermentable monomers.
  • yields were obtained of up to 35% on mole to mole glucose basis in shake flask tests. In these tests no process optimalization has taken place yet. With the AATs according to the invention yields may be reached of up to 60 % ethyl acetate on mole to mole glucose basis.
  • the polypeptides according to the invention are able to have a yield ranging from 10 to 55% mole ethyl acetate per mole glucose. In most cases, a yield of 45 to 55 % mole of ethyl acetate per mole of glucose may be reached.
  • the invention provides methods for producing an acetate ester which, in addition to the steps detailed above, further comprise the step of recovering the ethyl acetate. This comprises recovery of the ethyl acetate from the host cell mixture and optionally additional purification.
  • Suitable purification can be carried out by methods known to the person skilled in the art such as by using distillation, extraction, ion exchange resins, electrodialysis, nanofiltration, etc.
  • pYES2 derived plasmids were constructed by inserting genes into the multiple cloning site, either by using appropriate restriction enzymes, or by in vivo yeast assembly (Saccharomyces cerevisiae. Bio-protocol 5, (2015) pCUP1 derived plasmids were constructed by replacing the GAL1 promoter of pYES2 with the S. cerevisiae NCYC 2629 CUP1 promoter and inserting a gene of interest with in vivo yeast recombination, using S. cerevisiae W303 or CEN.PK2-1 D. S. cerevisiae transformations were performed according to the protocol of Gietz and Woods (Methods Enzymol.
  • pYES2 and pCUP1 derived plasmids were characterized in S. cerevisiae INVScl and CEN.PK2-1 D, respectively.
  • pET26b:harm AAT 1 - His was constructed by cloning the E. coli cod on harmonized AAT 1 gene with Ndel and Xhol in accordance with the method described in PLoS One 3, e2189 (2008).
  • ⁇ DE3 ⁇ sBamHIo AEcoRI-B int::(lacl::PlacUV5::T7gene1 ) 121 ⁇
  • pCUP1 :AAT 1 1 Non- S. cerevisiae, CUP1 IV. anomalus DSM 6766 producer) (WANOMALA 5545)
  • pCUP1 :AAT 12 Non- S. cerevisiae, AT. marxianus DSM 5422 producer
  • pCUP1 :AAT 14 Non- S. cerevisiae, C. fabianii CBS 5640 producer)
  • CUP1 CDR40570.1
  • pCUP1 :AAT 15 Non- S. cerevisiae, CUP1 S. cerevisiae NCYC 2629 IM032 producer) (YGR015C)
  • Wild type yeast strains were routinely cultured and propagated in YPD medium (20 g/L glucose, 20 g/L peptone, 10 g/L yeast extract).
  • Uracil auxotrophic yeast strains were routinely cultured and propagated in YS (yeast synthetic) medium (6.7 g/L Yeast nitrogen base without amino acids, 1.92 g/L Medium Supplements without uracil) with 20 g/L glucose.
  • E. co// ' strains were routinely cultured and propagated in LB or M9 medium supplemented with 50 pg/mL ampicillin or kanamycin. Yeast and E.
  • S. cerevisiae INVSd strains carrying pYES2 derived plasmids were characterized in 250 mL Erienmeyers containing 50 mL YS medium with 20 g/L galactose and 10 g/L raffinose. Erienmeyers were inoculated from an overnight culture and cultivated 48-72 hours at 250 rpm.
  • ciferrii CBS 1 1 1 were tested in 1 L Erienmeyers containing 250 mL YM (yeast minimal) medium (Can. J. Microbiol. 24, 440-447 (1978)). Iron was omitted from the medium. 1 ml_ 1000X vitamins mix (0.1 g/L thiamine, riboflavin, panthothenic acid, D-biotin, folic acid, p-aminobenzoic acid, cobalamin and 0.5 g/L nicotinic acid, pyridoxamine-HCI, pyridoxal-HCI) was added to the medium. Flasks were inoculated with 1 mL overnight YPD cultures and shaken at 150 rpm.
  • coli pre-cultures were made by inoculating 50 mL YS medium with 20 g/L glucose or 50 mL M9 medium, respectively, in a 250 mL Erlenmeyer flask to an initial OD of 0.5. The cultures were grown at 250 rpm and 30°C until an OD of 3. The cells were centrifuged at 4700 rpm for 5 minutes, resuspended in 50 mL sterile water and transferred to the fermenter. E. coli cultures were induced with 0.2 mM IPTG after 4 hours of growth.
  • the pH of the medium vessel was lowered to 2.0 by adding 37% HCI.
  • the dilution rate was 0.1 h ⁇ 1 .
  • Ethyl acetate production was controlled by aeration. Producing (aerobic) and non-producing (oxygen limited) conditions were achieved by sparging the fermenter with 3 L/h air or a 0.3 L/h air, 2.7 L/h N 2 mixture, respectively. Steady state was achieved after 50 h. 50 mL medium was withdrawn, centrifuged and the pellets frozen at -80 'C.
  • the pellets from the continous cultures were kept on ice as much as possible during the protocol.
  • the pellets were resuspended in 0.5 ml_ cold TE buffer (10 mM Tris-HCI, pH 8.0, 1 mM EDTA) and divided into two 2 ml_ screw-capped tubes containing 0.5 g zirconium beads, 30 ⁇ _ 10 %SDS, 30 ⁇ _ 3 M sodium acetate (pH 5.2) and 500 ⁇ _ Roti-® Phenol (pH 4.5-5.0, Roth).
  • the cells were disrupted with a FastPrep apparatus (ex MP biomedials) at speed 6 for 40 seconds and centrifuged at 4 °C and 14.000 rpm for 5 minutes.
  • RNAseq analysis was performed by BaseClear and the reads mapped to the annotated genome of W. anomalus DSM 6766. See FEMS Yeast Res. 12, 382-386 (2012).
  • wan5543 Homologs of wan5543 were identified by performing standard protein BLAST searches against the non-redundant protein sequences database. Phylogenetic analysis was performed by making a structural alignment of wan5543 and close homologues using the Tcoffee (Espresso) server (http:/7tcoffee.crg.cat/apps/tcoffee/do:expresso) [1 ]. A three-dimensional structure of Wan5543 was modeled using the PHYRE2 Protein Fold Recognition Server (http://www.sbg.bio.ic.ac.uk/phyre2) on intensive mode [2].
  • PHYRE2 Protein Fold Recognition Server http://www.sbg.bio.ic.ac.uk/phyre2
  • AAT 1 was purified from E. coli BL21 (DE3) pET26b:harmAAT 1 -His. Three 1 L Erlenmeyers with 250 mL M9 medium were inoculated with 2 mL overnight preculture and cultivated at 37 °C and 200 rpm. After 4 hours the cultures were chilled on ice for 15 minutes and induced with 0.2 mM IPTG. The cultures were then incubated at 20 °C and 200 rpm. After 18 hours, the cells were combined, harvested by centrifugation at 5000 rpm for 5 minutes, washed with 50 mL 50 mM phosphate buffer (KPi, pH 7.5) and stored at -20 °C.
  • KPi 50 mM phosphate buffer
  • the cells were resuspended in 20 mL buffer HA (50 mM KPi, 300 mM NaCI, pH 8) and passed twice through a chilled French Press Cell (Thermo Scientific) at 20000 psi. 25 mg DNAsel was added to the lysate, which was centrifuged at 4 °C and 18000 rpm for 20 minutes. The supernatant was filtered through 0.45 ⁇ filter and was used for protein purification. An AKTA Purifier system was used to purify AAT 1 . The cell-free extract was loaded on a 1 mL HisTrap HP column (GE Healthcare Life Sciences) that was equilibrated with buffer HA.
  • buffer HA 50 mM KPi, 300 mM NaCI, pH 8
  • 25 mg DNAsel was added to the lysate, which was centrifuged at 4 °C and 18000 rpm for 20 minutes. The supernatant was filtered through 0.45 ⁇ filter and was used for protein purification
  • the protein was eluted with a gradient of buffer HB (50 mM KPi, 300 mM NaCI, 500 mM imidazole, pH 8).
  • the fractions containing the protein were desalted over three connected 5 mL HiTrap Desalting columns (GE Healthcare Life Sciences), equilibrated with buffer CA (50 mM KPi, pH 7).
  • the desalted protein fractions were loaded on a 1 mL HiTrap HP SP column (GE Healthcare Life Sciences).
  • AAT 1 was eluted with a gradient of buffer CB (50 mM KPi, pH 7, 1 M NaCI). The fractions with the highest content of protein were combined and used for further analyses.
  • Glucose and organic acids were analyzed by HPLC using an ICS5000 HPLC system (Thermo Scientific) equipped with a Dionex DP pump, Dionex AS-AP autosampler, Dionex VWD UV detector operated at 210 nm and Shodex Rl detector operated at 35 °C.
  • An Aminex HPX-87H cation-exchange column was used with a mobile phase of 0.16 N H 2 S0 4 and was operated at 0.8 mL/min and 60°C. 10 mM dimethylsulfoxide in 0.04 N H 2 S0 4 was used as internal standard.
  • Volatile compounds were analyzed on a Shimadzu 2010 gas chromatograph equipped with a 20i- s autosampler.
  • the sequenced and annotated genome of ethyl acetate producing Wickerhamomyces anomalus DSM 6766 contains five putative Atf 1 or Atf2 homologs and one Eht1 homolog. To see if they are involved in ethyl acetate production they were expressed in S. cerevisiae INVSc (See Table I). The transformants 0.005 g/L ethyl acetate at most. However, they did produce 3 - 15 fold more isoamyl acetate from the endogenously produced isoamyl alcohol. Overexpression of the S. cerevisiae atfl gave similar results, but S. cerevisiae INVSd expressing atf2 and ehtl showed poor growth and did not produce esters.
  • the known homologs of Atf 1 , Atf2 and Eht1 did not show a significant change in expression levels.
  • the overexpressed genes that were not upregulated due to the metabolic shift from an aerobic to fermentative metabolism during ethyl acetate production code for two hypothetical proteins with an alpha/beta hydrolase fold. This fold has been observed in esterases, as well as some AATs, see J. Biol. Chem. 281 , 4446-4456 (2006). Both types of enzymes are involved in ester metabolism in yeast, making AAT 1 and WANOMALA 7754 potential candidates for explaining ethyl acetate formation in W. anomalus. Their protein products are 99% identical and only AAT 1 was studied further.
  • the enzyme was expressed in S. cerevisiae INVSd pYES2-AAT 1 , which produced 0.13 ⁇ 0.01 g/L ethyl acetate. This is 26-fold higher than the best W. anomalus Atf 1 , Atf2 or Eht1 homolog.
  • S. cerevisiae INVSd was grown on galactose, which also served as the inducer for gene expression.
  • Codon harmonized AAT 1 was expressed in E. coli BL21 (DE3) pET26b:harm AAT 1 -His. This led to the peak production of 3.5 ⁇ 0.12 g/L ethyl acetate from 20 g/L glucose and 15 g/L ethanol. E. coli produced 29-fold more ethyl acetate than S. cerevisiae (26 % of the theoretical pathway maximum).
  • Example 3 Biochemical characterization of AAT 1 Automatic annotation predicted an ⁇ / ⁇ hydrolase fold in AAT1. Based on this we assumed that AAT 1 makes ethyl acetate either as an AAT or as a reversed esterase, and excluded HADH.
  • AAT 1 was purified from E. coli BL21 (DE3) pET26b:harm AAT 1 -His by Ni/NTA affinity chromatography, followed by cation exchange. The purified AAT1 protein fraction was subjected to a GC assay for AAT activity. To this end both ethanol and acetyl coenzyme A were contacted with AAT1. When omitting either ethanol and/or acetyl coenzyme A no ethyl acetate was formed. This confirms that AAT 1 produces ethyl acetate as an AAT, and not as a reversed esterase.
  • the AAT 1 homologs used to compile the multiple sequence alignment originate from other ethyl acetate-producing yeast species.
  • CEN.PK2- 1 D several of the transformants produced ethyl acetate upon expression in a host.
  • the growth and product production profiles of the transformants resembled that of CEN.PK2-1 D pCUP1 :AAT 1 , but the final titer of ethyl acetate varied per homolog.
  • the amino acid sequences and the nucleotide sequences of the homologs that proved to have ethyl acetate production activity are compiled in sequence listings.
  • At least one ethyl acetate producing homolog of AAT 1 was present in each ethyl acetate-producing yeast.
  • the homologs found which proved to have ethyl acetate producing activity were genes isolated from Kluyveromyces marxianus, Kluyveromyces lactis, Wickerhamomyces ciferrii, Cyberlindnera jadinii Hanseniaspora u varum, Eremothecium cymbalariae.and S. cerevisiae. These genes showed roughly 50% identity to AAT 1 .
  • These new enzymes are referred to as AAT 2 to AAT 10.
  • amino sequences of these AAT 2-10 are given under SEQ ID No:3, SEQ ID No:5, SEQ ID No:7, SEQ ID No:9, SEQ ID No:1 1 , SEQ ID No:13, SEQ ID No:15, SEQ ID No:17 and SEQ ID No: 19, respectively.
  • the nucleotide sequences encoding for AAT 2-10 are given in SEQ ID NO: 4, SEQ ID No: 6, SEQ ID NO: 8, SEQ ID No: 10, SEQ ID NO: 12, SEQ ID No: 14, SEQ ID NO: 16, SEQ ID No: 18, and SEQ ID NO: 20, respectively.
  • Non-producers sequences of the polypeptides that proved to have ethyl acetate production activity (such as in AAT 1 to AAT 10, hereinafter referred to as Producers) and sequences of polypeptides that did not prove to have ethyl acetate production activity (hereinafter referred to as Non-producers), it was found that in the Producers, the histidine of the serine- aspartic acid-histidine triad or serine-glycine-histidine triad was present in a polypeptide fragment that has a glutamic acid-arginine-proline (ERP) fragment, a glutamic acid-asparagine-proline (ENP) fragment or glutamic-acid-methionine-proline (EMP) fragment as the 5 th , 6 m and 7 th amino acid from the histidine on the C-terminal side of the polypeptide.
  • the polypeptides that turned out to be a Non-producer did not
  • AAT 1 homologs were also present in Cyberlindnera fabianii CBS 5640 and Wickerhamomyces ciferrii CBS 1 1 1 (AAT 6 and AAT 2, respectively). To our knowledge, these strains have not been studied for ethyl acetate production before. Therefore, their potential for ethyl acetate production was tested in shake flasks in YM medium. C. fabianii and W. ciferrii produced 4.1 and 5.7 g/L ethyl acetate from 50 g/L glucose, respectively.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

La présente invention concerne un procédé de production d'acétate d'éthyle dans lequel un polypeptide ayant une activité d'alcool acétyle transférase convertit l'éthanol et l'acétyle coenzyme A en acétate d'éthyle. Le polypeptide possède un repliement alpha-bêta hydrolase et un site actif comprenant une sérine, de l'histidine et éventuellement de l'acide aspartique dans sa structure tridimensionnelle. Les polypeptides selon l'invention sont des gènes isolés à partir de Kluyveromyces marxianus, Kluyveromyces lactis, Wickerhamomyces ciferrii, Cyberlindnera jadinii, Hanseniaspora uvarum, Eremothecium cymbalarie et Saccharomyces cerevisiae.
PCT/EP2017/081048 2016-12-01 2017-11-30 Alcool acétyl-transférases pour la production d'acétate d'éthyle WO2018100097A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP16201644.8 2016-12-01
EP16201644 2016-12-01

Publications (1)

Publication Number Publication Date
WO2018100097A1 true WO2018100097A1 (fr) 2018-06-07

Family

ID=57544202

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/081048 WO2018100097A1 (fr) 2016-12-01 2017-11-30 Alcool acétyl-transférases pour la production d'acétate d'éthyle

Country Status (1)

Country Link
WO (1) WO2018100097A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115197858A (zh) * 2022-08-02 2022-10-18 劲牌有限公司 一种高产乙酸乙酯费比恩毕赤酵母在清香型小曲白酒中的应用
CN115466686A (zh) * 2021-09-14 2022-12-13 安琪酵母股份有限公司 一种费比恩毕赤酵母菌株及其应用
KR20240086789A (ko) 2022-12-06 2024-06-19 중앙대학교 산학협력단 위커하모마이세스 서브펠리큐로서스 유래 신규 에탄올 아세틸트랜스퍼레이즈 및 이를 이용한 에틸 아세테이트 생산방법

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102199556B (zh) * 2011-04-15 2013-03-27 天津科技大学 一种高产酯酿酒酵母基因工程菌及其构建方法
US20130295616A1 (en) 2011-01-20 2013-11-07 Toyota Jidosha Kabushiki Kaisha Recombinant yeast and substance production method using the same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130295616A1 (en) 2011-01-20 2013-11-07 Toyota Jidosha Kabushiki Kaisha Recombinant yeast and substance production method using the same
CN102199556B (zh) * 2011-04-15 2013-03-27 天津科技大学 一种高产酯酿酒酵母基因工程菌及其构建方法

Non-Patent Citations (26)

* Cited by examiner, † Cited by third party
Title
"Current Protocols in Molecular Biology", 1995, JOHN WILEY & SONS
ANTHONY R. BORNEMAN ET AL: "Whole-Genome Comparison Reveals Novel Genetic Elements That Characterize the Genome of Industrial Strains of Saccharomyces cerevisiae", PLOS GENETICS, vol. 7, no. 2, 3 February 2011 (2011-02-03), pages e1001287, XP055067199, DOI: 10.1371/journal.pgen.1001287 *
APPLIED MICROBIOL BIOTECHNOL, vol. 98, 2014, pages 5397 - 5415
ARMOUGOM F; MORETTI S; POIROT O; AUDIC S; DUMAS P; SCHAELI B; KEDUAS V; NOTREDAME C, NUCLEIC ACIDS RES., 2006
CAN. J. MICROBIOL., vol. 24, 1978, pages 440 - 447
DATABASE UniProt [online] 14 October 2015 (2015-10-14), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:CEP25158.1};", XP055391264, retrieved from EBI accession no. UNIPROT:A0A0H5C9V5 Database accession no. A0A0H5C9V5 *
DATABASE UniProt [online] 16 August 2004 (2004-08-16), "SubName: Full=KLLA0E24421p {ECO:0000313|EMBL:CAH00138.1};", XP055391265, retrieved from EBI accession no. UNIPROT:Q6CLY8 Database accession no. Q6CLY8 *
DATABASE UniProt [online] 19 March 2014 (2014-03-19), "SubName: Full=Uncharacterized abhydrolase domain-containing protein YGR015C {ECO:0000313|EMBL:BAO38427.1};", XP055391257, retrieved from EBI accession no. UNIPROT:W0T4A7 Database accession no. W0T4A7 *
DATABASE UniProt [online] 24 June 2015 (2015-06-24), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:KKA02732.1};", XP055391339, retrieved from EBI accession no. UNIPROT:A0A0F4X9N5 Database accession no. A0A0F4X9N5 *
DATABASE UniProt [online] 24 June 2015 (2015-06-24), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:KKA03697.1};", XP055391279, retrieved from EBI accession no. UNIPROT:A0A0F4XCF2 Database accession no. A0A0F4XCF2 *
DATABASE UniProt [online] 25 January 2012 (2012-01-25), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:AET40929.1};", XP055391343, retrieved from EBI accession no. UNIPROT:G8JVR4 Database accession no. G8JVR4 *
DATABASE UniProt [online] 28 November 2012 (2012-11-28), "SubName: Full=Abhydrolase domain-containing protein 11 {ECO:0000313|EMBL:CCH45056.1}; EC=3.-.-.- {ECO:0000313|EMBL:CCH45056.1};", XP055391246, retrieved from EBI accession no. UNIPROT:K0KPV8 Database accession no. K0KPV8 *
DATABASE UniProt [online] 3 September 2014 (2014-09-03), "SubName: Full=Abhydrolase domain-containing protein IMO32 {ECO:0000313|EMBL:ONH67986.1}; SubName: Full=CYFA0S05e02014g1_1 {ECO:0000313|EMBL:CDR40574.1};", XP055391272, retrieved from EBI accession no. UNIPROT:A0A061AYY2 Database accession no. A0A061AYY2 *
DATABASE UniProt [online] 5 April 2011 (2011-04-05), "SubName: Full=YGR015C-like protein {ECO:0000313|EMBL:EGA82805.1};", XP055391346, retrieved from EBI accession no. UNIPROT:E7KNL2 Database accession no. E7KNL2 *
FEMS YEAST RES., vol. 12, 2012, pages 382 - 386
GIETZ; WOODS, METHODS ENZYMOL., vol. 350, 2002, pages 87 - 96
HOLMQUIST M: "Alpha/Beta-hydrolase fold enzymes: structures, functions and mechanisms", CURRENT PROTEIN AND PEPTIDE SCI, BENTHAM SCIENCE PULBISHERS, NL, vol. 1, no. 2, 1 September 2000 (2000-09-01), pages 209 - 235, XP009126096, ISSN: 1389-2037 *
J. BIOL. CHEM., vol. 281, 2006, pages 4446 - 4456
KELLEY LA ET AL., NATURE PROTOCOLS, vol. 10, 2015, pages 845 - 858
KNIGHT, M. J.; BULL, I. D.; CURNOW, P., YEAST, vol. 31, 2014
MICROB. BIOTECHNOL., vol. 3, 2010, pages 165 - 177
NEEDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 444 - 453
PLOS ONE, vol. 3, 2008, pages e2189
SAMBROOK ET AL.: "Molecular Cloning, A Laboratory Manual", 1989, COLD SPRING HARBOR PRESS
YEAST, vol. 21, 2004, pages 781 - 792
YEAST, vol. 8, 1992, pages 501 - 517

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115466686A (zh) * 2021-09-14 2022-12-13 安琪酵母股份有限公司 一种费比恩毕赤酵母菌株及其应用
CN115197858A (zh) * 2022-08-02 2022-10-18 劲牌有限公司 一种高产乙酸乙酯费比恩毕赤酵母在清香型小曲白酒中的应用
CN115197858B (zh) * 2022-08-02 2023-05-05 劲牌有限公司 一种高产乙酸乙酯费比恩毕赤酵母在清香型小曲白酒中的应用
KR20240086789A (ko) 2022-12-06 2024-06-19 중앙대학교 산학협력단 위커하모마이세스 서브펠리큐로서스 유래 신규 에탄올 아세틸트랜스퍼레이즈 및 이를 이용한 에틸 아세테이트 생산방법

Similar Documents

Publication Publication Date Title
Tang et al. Microbial conversion of glycerol to 1, 3-propanediol by an engineered strain of Escherichia coli
KR100679638B1 (ko) 포메이트 디하이드로게나제 d 또는 e를 코딩하는 유전자로 형질전환된 미생물 및 이를 이용한 숙신산의 제조방법
US9909146B2 (en) Production of alkenes by enzymatic decarboxylation of 3-hydroxyalkanoic acids
RU2609656C2 (ru) Способ получения алкенов путем комбинированного ферментативного превращения 3-гидроксиалкановых кислот
EP3325608B1 (fr) Procédés et micro-organismes de production de 1,3-butanediol
US20130095542A1 (en) Engineered microorganisms and integrated process for producing n-propanol, propylene and polypropylene
Kwak et al. Biosynthesis of 3-hydroxypropionic acid from glycerol in recombinant Escherichia coli expressing Lactobacillus brevis dhaB and dhaR gene clusters and E. coli K-12 aldH
US20150010968A1 (en) Biological alkene oxidation
WO2010022763A1 (fr) Procédé de préparation de 2-hydroxy-isobutyrate
WO2012109534A2 (fr) Cellules et procédés de production d'acide isobutyrique
US20150299741A1 (en) Method for conversion of an alkane or 1-alkanol to a diol
US20110014666A1 (en) Polypeptide having glyoxalase iii activity, polynucleotide encoding the same and uses thereof
US20120301935A1 (en) Recombinant microorganism for simultaneously producing 3-hydroxypropionic acid and 1,3 propanediol
JP4809660B2 (ja) 3−キヌクリジノン還元酵素およびこれを用いる(r)−3−キヌクリジノールの製造方法
WO2018100097A1 (fr) Alcool acétyl-transférases pour la production d'acétate d'éthyle
CN114026246A (zh) 从可再生来源生产化学品
CN113416686B (zh) 具有增加的碳通量效率的方法和生物
CN110396507B (zh) 源自Cnuibacter physcomitrellae的L-泛解酸内酯脱氢酶
JP2017534268A (ja) 有用産物の生産のための改変微生物および方法
Jang et al. Whole cell biotransformation of 1-dodecanol by Escherichia coli by soluble expression of ADH enzyme from Yarrowia lipolytica
WO2018099719A1 (fr) Alcool-acétyl transférases pour la production d'alcanoate d'alkyle
Radoš et al. Stereospecificity of Corynebacterium glutamicum 2, 3-butanediol dehydrogenase and implications for the stereochemical purity of bioproduced 2, 3-butanediol
US9637761B2 (en) Recombinant microorganism metabolizing 3,6-anhydride-L-galactose and a use thereof
CN110527671B (zh) 源自Nocardia farcinica的L-泛解酸内酯脱氢酶及其应用
CN115896065A (zh) 一种立体选择性羧酯酶、编码基因、载体及其应用

Legal Events

Date Code Title Description
DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17816586

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17816586

Country of ref document: EP

Kind code of ref document: A1