MATERIALS AND METHODS FOR BIOSYNTHETIC MANUFACTURE AND UTILIZATION OF SYNTHETIC POLYPEPTIDES, AND PRODUCTS THEREFROM

Information

  • Patent Application
  • 20190359957
  • Publication Number
    20190359957
  • Date Filed
    April 01, 2019
    5 years ago
  • Date Published
    November 28, 2019
    4 years ago
Abstract
Provided herein are novel, synthetic polypeptides having, for example, acyl-acyl carrier protein (ACP) thioesterase (TE) activity, including polypeptides that convert pimeloyl-ACP to pimelic acid. In some aspects, the synthetic polypeptides have advantageous enzymatic activity and/or improved substrate specificity relative to a wild type acyl-ACP TE.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been filed electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jul. 24, 2019, is named 061646-1131147_(00601US)_SL.txt and is 366,159 bytes in size.


FIELD

The teachings in the present disclosure relate to novel, synthetic polypeptides having, for example, acyl-acyl carrier protein (ACP) thioesterase (TE) activity, including polypeptides that convert pimeloyl-ACP to pimelic acid. In some aspects of the present disclosure, the synthetic polypeptides have advantageous enzymatic activity and/or improved substrate specificity relative to a wild type acyl-ACP TE.


BACKGROUND

Biochemical pathways are utilized to produce products useful for an organism. See, for example, LEHNINGER, A. L., NELSON, D. L., & COX, M. M. (2000). Lehninger principles of biochemistry. New York, Worth Publishers. A biochemical pathway for the production of 7-Aminoheptanoic acid (7-AHA) utilizes the biotin biosynthesis pathway in E. coli (Lin et al., “Biotin Synthesis Beings by Hijacking the Fatty Acid Synthesis Pathway,” Nat. Chem. Biol. 6(9): 682-688 2010). The first committed step in biotin biosynthesis is the methylation of malonyl-ACP by BioC. The malonyl-ACP methyl ester thus generated then serves as a starter unit for the fatty acid biosynthesis pathway.


Two rounds of fatty acid elongation and reduction then occur to generate pimeloyl-ACP methyl ester. Pimeloyl-ACP is then generated by the removal of the methyl group by the esterase BioH (FIG. 1). Pimeloyl-ACP is then acted upon by a thioesterase enzyme to produce pimelic acid. Thioesterases (TE) therefore play a crucial role in the production of 7-AHA using the biotin synthesis pathway (FIG. 2).


Polyamides, such as nylons, can be synthesized by the condensation polymerisation of a diamine with a dicarboxylic acid or alternatively by the condensation polymerisation of lactams. Nylon 6,6 is a ubiquitous nylon produced by the reaction of hexamethylenediamine (HMD) and adipic acid. Nylon 6 is produced by a ring opening polymerisation of caprolactam. Nylon 7 represents a novel polyamide with value-added characteristics compared to Nylon 6 and Nylon 6,6. The key intermediate for the synthesis of Nylon 7 is 7-AHA. However, no economically favorable petrochemical routes exist to produce 7-AHA.


BRIEF SUMMARY

Synthetic biology is exploited for the design and construction of new biological parts, devices, and systems, and the re-design of existing, natural biological systems for useful purposes. In one aspect, synthetic polypeptides, having an acyl-ACP TE activity, useful for the production of 7-AHA, via biosynthetic routes, are taught.


The present disclosure, in one embodiment, provides various polypeptides having acyl-ACP TE activity. In another aspect, the polypeptides having acyl-ACP TE activity comprise one or more amino acid substitution(s) relative to a wild-type acyl-ACP TE, wherein the one or more amino acid substitution(s) are at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1, or a functional fragment thereof.


In some embodiments, the wild-type acyl-ACP TE is classified under EC 3.1.2.-. In some embodiments, the wild-type acyl-ACP TE is classified under EC 3.1.2.14.


In some embodiments, the amino acid corresponding to position 5 is substituted with tyrosine (Y) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 35 is substituted with serine (S) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 38 is substituted with glutamine (Q) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 64 is substituted with valine (V) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 241 is substituted with glutamic acid (E) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 45 is substituted with methionine or isoleucine (I) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 128 is substituted with tyrosine (Y) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 175 is substituted with serine (S) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 33 is substituted with aspartic acid (D) or an equivalent amino acid, and the amino acid corresponding to position 128 is substituted with tyrosine (Y) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 59 is substituted with valine (V) or an equivalent amino acid, and the amino acid corresponding to position 90 is substituted with phenylalanine (F) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 40 is substituted with glutamic acid (E) or an equivalent amino acid, and the amino acid corresponding to position 111 is substituted with tryptophan (W) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 36 is substituted with glycine (G) or an equivalent amino acid, and the amino acid corresponding to position 128 is substituted with tyrosine (Y) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 32 is substituted with glutamine (Q) or an equivalent amino acid, and the amino acid corresponding to position 40 is substituted with glutamic acid (E) or an equivalent amino acid.


In some embodiments, the amino acid sequence of the polypeptide having an acyl-ACP TE activity has at least 50% amino acid sequence identity with the amino acid sequence of the wild-type acyl-ACP TE.


In some embodiments, the polypeptide having an acyl-ACP TE activity has increased enzymatic activity and/or improved substrate specificity relative to the wild-type acyl-ACP TE. In certain embodiments the the polypeptide having an acyl-ACP TE activity has improved substrate specificity for pimeloyl-ACP relative to the wild-type acyl-ACP TE. In certain embodiments, the the polypeptide having an acyl-ACP TE activity has an increase in enzymatic activity of at least 10% relative to the wild-type acyl-ACP TE.


The present disclosure provides, in another embodiment, a nucleic acid encoding a polypeptide having an acyl-ACP TE activity. In some embodiments, the nucleic acid encodes a polypeptide having an acyl-ACP TE activity, wherein the polypeptide comprises one or more amino acid substitution(s) relative to a wild-type acyl-ACP TE, wherein the one or more amino acid substitution(s) are at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1, or a functional fragment thereof. Also provided, in another embodiment, is a vector comprising a nucleic acid encoding a polypeptide having acyl-ACP TE activity. In some embodiments, the vector comprises a nucleic acid encoding a polypeptide having an acyl-ACP TE activity, wherein the polypeptide comprises one or more amino acid substitution(s) relative to a wild-type acyl-ACP TE, wherein the one or more amino acid substitution(s) are at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1.


Further provided, in another embodiment, is a metabolically engineered organism or a host cell comprising any of the vectors provided herein. In some embodiments, the host cell is a prokaryotic cell.


Also provided, in another embodiment, is a composition comprising any of the polypeptides described herein. In some embodiments, the composition further comprises pimeloyl-ACP.


Further provided, in another embodiment, is a method for producing pimelic acid, pimelate semialdehyde or 7-AHA, the method comprising the step of converting pimeloyl-ACP to pimelic acid in the presence of any one of the polypeptides provided herein. Also, provided, in another embodiment, is a method for producing pimelic acid, pimelate semialdehyde or 7-AHA, the method comprising the steps of culturing a host cell comprising a vector encoding any of the polypeptides described herein in a suitable medium and recovering the pimelic acid, pimelate semialdehyde or 7-AHA.


Also provided, in another embodiment is a bioderived pimelic acid, pimelate semialdehyde or 7-AHA, that is produced by any of the methods provided herein. Further provided, in another embodiment, is a product comprising a chemical produced from any of the bioderived products provided herein, wherein the product comprises a pharmaceutical, a biofuel, a fragrance, a nylon intermediate, a polyester, or a food additive.


Also provided is a bio-based or fermentation-derived product, wherein said product comprises: (i) a composition comprising at least one bio-derived, bio-based or fermentation-derived compound, or salts thereof, produced in the presence any of the polypeptides described herein, or produced from the product of a reaction catalyzed by any of the polypeptides described herein, or any combination thereof; (ii) a bio-derived, bio-based or fermentation-derived polymer comprising the bio-derived, bio-based or fermentation-derived composition or compound of (i), or any combination thereof (iii) a bio-derived, bio-based or fermentation-derived resin comprising the bio-derived, bio-based or fermentation-derived compound or bio-derived, bio-based or fermentation-derived composition of (i), or any combination thereof or the bio-derived, bio-based or fermentation-derived polymer of (ii), or any combination thereof (iv) a substance obtained by molding the bio-derived, bio-based or fermentation-derived polymer of (ii), or the bio-derived, bio-based or fermentation-derived resin of (iii), or any combination thereof (v) a bio-derived, bio-based or fermentation-derived formulation comprising the bio-derived, bio-based or fermentation-derived composition of (i), bio-derived, bio-based or fermentation-derived compound of (i), bio-derived, bio-based or fermentation-derived polymer of (ii), bio-derived, bio-based or fermentation-derived resin of (iii), or bio-derived, bio-based or fermentation-derived substance of (iv), or any combination thereof, or; (vi) a bio-derived, bio-based or fermentation-derived semi-solid or a non-semi-solid stream, comprising the bio-derived, bio-based or fermentation-derived composition of (i), bio-derived, bio-based or fermentation-derived compound of (i), bio-derived, bio-based or fermentation-derived polymer of (ii), bio-derived, bio-based or fermentation-derived resin of (iii), bio-derived, bio-based or fermentation-derived formulation of (v), bio-derived, bio-based or fermentation-derived molded substance of (iv), or any combination thereof.





BRIEF DESCRIPTION OF THE FIGURES

The present application includes the following figures. The figures are intended to illustrate certain embodiments and/or features of the compositions and methods, and to supplement any description(s) of the compositions and methods. The figures do not limit the scope of the compositions and methods, unless the written description expressly indicates that such is the case.



FIG. 1 is a schematic of a biochemical pathway for the production of pimeloyl-ACP from malonyl-ACP.



FIG. 2 is a schematic of a biochemical pathway for producing 7-AHA from malonyl-CoA.



FIG. 3 depicts a three-dimensional model of the Acyl-ACP TE obtained from Uniprot ID No. R5FQ35 highlighting catalytic residues.



FIG. 4A shows site-specific distribution of residues in the acyl-ACP TEs. A population of more than 40% was considered.



FIG. 4B shows the major contribution of amino acid residues 45, 128 and 175 at the binding site. Residue numbering used in this study was obtained from the sequence with Uniprot ID R5FQ35.



FIG. 5 shows the correlated residues at the substrate binding site for higher activity of Acyl-ACP-TEs. The degree of correlation is plotted as a heat-map (left panel) and the top 5 correlated residues are shown in the table (center panel). The correlated residues indicate that these two positions are coupled, and favours a residue at position two when a specific residue is present at position one. Spatial occupancy of the correlated residues is shown as a sphere in the 3D model of the enzyme (right panel).





DETAILED DESCRIPTION

All references cited herein and the material for which they are cited are hereby specifically incorporated by reference in their entireties.


Many modifications and other embodiments of the disclosures set forth herein will come to mind to one skilled in the art to which these disclosures pertain having the benefit of the teachings presented in the foregoing and following descriptions and the associated drawings. Therefore, it is to be understood that the disclosures are not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. Although specific terms are employed herein, they are used in a generic and descriptive sense only and not for purposes of limitation.


Units, prefixes and symbols may be denoted in their SI accepted form. Unless otherwise indicated, nucleic acids are written left to right in 5′ to 3′ orientation; amino acid sequences are written left to right in amino to carboxy orientation, respectively. Numeric ranges are inclusive of the numbers defining the range. Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes. The terms defined below are more fully defined by reference to the specification as a whole.


In the present description and claims, the conventional one-letter and three-letter codes for amino acid residues are used. All of the numbering, including the numbering of the substitutions is with reference to corresponding amino acid residues in the amino acid sequence set forth herein as SEQ ID NO: 1. SEQ ID NO: 1 is the full-length amino acid sequence for an Eggerthella sp. acyl-ACP TE deposited under Uniprot ID No. R5FQ35. In the present description and claims, the activity of the claimed polypeptide is measured relative to that of a wild-type acyl-ACP TE, unless otherwise specified.


“Correspondence” to another sequence (e.g., regions, fragments, nucleotide or amino acid positions, or the like), for example, SEQ ID NO: 1, is based on the convention of numbering according to nucleotide or amino acid position number and then aligning the sequences in a manner that maximizes the percentage of sequence identity. Because not all positions within a given “corresponding region” need be identical, non-matching positions within a corresponding region may be regarded as “corresponding positions.”


Definitions

Unless specifically defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Unless mentioned otherwise, the techniques employed or contemplated herein are standard methodologies well known to one of ordinary skill in the art. The practice of the present disclosure will employ, unless otherwise indicated, conventional techniques of microbiology, tissue culture, molecular biology, chemistry, biochemistry and recombinant DNA technology, which are within the skill of the art. The materials, methods and examples are illustrative only and not limiting. The following is presented by way of illustration and is not intended to limit the scope of the disclosure.


In some embodiments, numbers expressing quantities of reagents, properties, reaction conditions and results, and so forth, used to describe and claim certain embodiments of the present disclosure are to be understood as being modified in some instances by the term “about,” In some embodiments, the numerical parameters set forth in the specification (into which the claims are incorporated in their entirety) are approximations that can vary depending upon the desired properties sought to be obtained by a particular embodiment. In some embodiments, the numerical parameters should be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of some embodiments of the present disclosure are approximations, the numerical values set forth in the specific examples are reported as precisely as practical. The numerical values presented in some embodiments of the present disclosure may contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements. The recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein.


As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise.


An “amino acid” refers to any monomer unit that can be incorporated into a peptide, polypeptide, or protein. As used herein, the term “amino acid” includes the following twenty natural or genetically encoded alpha-amino acids: alanine (Ala or A), arginine (Arg or R), asparagine (Asn or N), aspartic acid (Asp or D), cysteine (Cys or C), glutamine (Gln or Q), glutamic acid (Glu or E), glycine (Gly or G), histidine (His or H), isoleucine (Ile or I), leucine (Leu or L), lysine (Lys or K), methionine (Met or M), phenylalanine (Phe or F), proline (Pro or P), serine (Ser or S), threonine (Thr or T), tryptophan (Trp or W), tyrosine (Tyr or Y), and valine (Val or V). The structures of these twenty natural amino acids are shown in, e.g., Stryer et al., Biochemistry, 5th ed., Freeman and Company (2002). The term “amino acid” also includes unnatural amino acids, modified amino acids (e.g., having modified side chains and/or backbones), and amino acid analogs.


As used herein, the term “bioderived” means derived from or synthesized by a biological organism and can be considered a renewable resource since it can be generated by a biological organism. Such a biological organism, in particular the microbial organisms disclosed herein, can utilize feedstock or biomass, such as, sugars or carbohydrates obtained from an agricultural, plant, bacterial, or animal source. Alternatively, the biological organism can utilize atmospheric carbon. As used herein, the term “biobased” means a product as described above that is composed, in whole or in part, of a bioderived compound of the disclosure. A biobased or bioderived product is in contrast to a petroleum derived product, wherein such a product is derived from or synthesized from petroleum or a petrochemical feedstock.


For compounds containing carboxylic acid groups such as organic monoacids, hydroxyacids, amino acids and dicarboxylic acids, these compounds may be formed or converted to their ionic salt form when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base. Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like. Acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. The salt can be isolated as is from the system as the salt or converted to the free acid by reducing the pH to below the pKa through addition of acid or treatment with an acidic ion exchange resin.


For compounds containing amine groups such as but not limited to organic amines, amino acids and diamine, these compounds may be formed or converted to their ionic salt form by addition of an acidic proton to the amine to form the ammonium salt, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene-1-carboxylic acid, glucoheptonic acid, 4,4′-methylenebis-(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid or muconic acid. Acceptable inorganic bases are known in the art and include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. The salt can be isolated as is from the system as a salt or converted to the free amine by raising the pH to above the pKb through addition of base or treatment with a basic ion exchange resin


For compounds containing both amine groups and carboxylic acid groups such as but not limited to amino acids, these compounds may be formed or converted to their ionic salt form by either 1) forming acid addition salts with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene-1-acid), carboxylic acid, glucoheptonic acid, 4,4′-methylenebis-(3-hydroxy-2-ene-1-carboxylic 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid. Acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like or 2) replacing an acidic proton present in the parent compound with a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base. Acceptable organic bases are known in the art and include ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like. Acceptable inorganic bases are known in the art and include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. The salt can be isolated as is from the system or converted to the free acid by reducing the pH to below the pKa through addition of acid or treatment with an acidic ion exchange resin.


The terms “comprise,” “have” and “include” are open-ended linking verbs. Any forms or tenses of one or more of these verbs, such as “comprises,” “comprising,” “has,” “having,” “includes” and “including,” are also open-ended. For example, any method that “comprises,” “has” or “includes” one or more steps is not limited to possessing only those one or more steps and can also cover other unlisted steps. Similarly, any composition that “comprises,” “has” or “includes” one or more features is not limited to possessing only those one or more features and can cover other unlisted features. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g. “such as”) provided with respect to certain embodiments herein is intended merely to better illuminate the present disclosure and does not pose a limitation on the scope of the present disclosure otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the present disclosure.


As used herein, “consisting essentially of” means the inclusion of additional sequences in a polynucleotide or polypeptide sequence provided herein where the additional sequences do not materially affect the basic function of the claimed polynucleotide or polypeptide sequences. With regard to compositions in general, the term “consisting essentially of” refers to those elements required for a given embodiment and additionally permits the presence of elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment of the disclosure.


As used herein, the term “consisting of” refers to compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.


The term “conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refer to those nucleic acids that encode identical or conservatively modified variants of the amino acid sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are “silent variations” and represent one species of conservatively modified variation. Every nucleic acid sequence herein that encodes a polypeptide also describes every possible silent variation of the nucleic acid. One of ordinary skill will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine; one exception is Micrococcus rubens, for which GTG is the methionine codon (Ishizuka, et al., (1993) J. Gen, Microbiol. 139:425-32) can be modified to yield a functionally identical molecule. Accordingly, each silent variation of a nucleic acid, which encodes a polypeptide of the present disclosure, is implicit in each described polypeptide sequence and incorporated herein by reference.


As to amino acid sequences, one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide or protein sequence which alters, adds, or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” when the alteration results in the substitution of an amino acid with a chemically similar amino acid. Thus, any number of amino acid residues can be so altered. Conservatively modified variants typically provide equivalent biological activity as the unmodified polypeptide sequence from which they are derived. Conservative substitution tables providing functionally similar amino acids, also referred herein as “equivalent amino acids” are well known in the art.


As used herein, “codon optimization” is the process of modifying a nucleotide sequence in a manner that improves its expression, G/C content, RNA secondary structure, and translation in eukaryotic cells, without altering the amino acid sequence it encodes. Altered codon usage is often employed to alter translational efficiency and/or to optimize the coding sequence for expression in a desired host or to optimize the codon usage in a heterologous sequence for expression in a particular host. Codon usage in the coding regions of the polynucleotides of the present disclosure can be analyzed statistically using commercially available software packages such as “Codon Preference” available from the University of Wisconsin Genetics Computer Group. See, Devereaux, et al., (1984) Nucleic Acids Res. 12:387-395) or MacVector 4.1 (Eastman Kodak Co., New Haven, Conn.). Thus, the present disclosure provides a codon usage frequency characteristic of the coding region of at least one of the polynucleotides of the present disclosure. The number of polynucleotides (3 nucleotides per amino acid) that can be used to determine a codon usage frequency can be any integer from 3 to the number of polynucleotides of the present disclosure as provided herein. Optionally, the polynucleotides will be full-length sequences. An exemplary number of sequences for statistical analysis can be at least 1, 5, 10, 20, 50 or 100.


The term “derived” encompasses the terms “originated from”, “obtained” or “obtainable from”, and “isolated from”.


“Equivalent amino acids” can be determined either on the basis of their structural homology with the amino acids for which they are substituted or on the results of comparative tests of biological activity between the various variants likely to be generated. As a non-limiting example, the list below summarizes possible substitutions often likely to be carried out without resulting in a significant modification of the biological activity of the corresponding variant:


1) Alanine (A), Serine (S), Threonine (T), Valine (V), Glycine (G), and Proline (P);


2) Aspartic acid (D), Glutamic acid (E);


3) Asparagine (N), Glutamine (Q);


4) Arginine (R), Lysine (K), Histidine (H);


5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V) and


6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W).


See also, Creighton, Proteins, W.H. Freeman and Co. (1984).


In making such changes/substitutions, the hydropathic index of amino acids may also be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle; (1982) J Mol Biol. 157(1):105-32). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors; DNA, antibodies, antigens and the like.


It is known in the art that certain amino acids may be substituted by other amino acids having a similar hydropathic index or score and still result in a protein with similar biological activity, i.e., still obtain a biological functionally equivalent protein. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics (Kyte and Doolittle, ibid). These are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (−0.4); threonine (−0.7); serine (−0.8); tryptophan (−0.9); tyrosine (−1.3); proline (−1.6); histidine (−3.2); glutamate (−3.5); glutamine (−3.5); aspartate (−3.5); asparagine (−3.5); lysine (−3.9) and arginine (−4.5). In making such changes, the substitution of amino acids whose hydropathic indices are within +2 is preferred; those which are within +1 are particularly preferred and those within +0.5 are even more particularly preferred.


It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity. U.S. Pat. No. 4,554,101, states that the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with a biological property of the protein.


As detailed in U.S. Pat. No. 4,554,101, the following hydrophilicity values have been assigned to amino acid residues; arginine (+3.0); lysine (+3.0); aspartate (+3.0.+0.1); glutamate (+3.0.+0.1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (−0.4); proline (−0.5.+0.1); alanine (−0.5); histidine (−0.5); cysteine (−1.0); methionine (−1.3); valine (−1.5); leucine (−1.8); isoleucine (−1.8); tyrosine (−2.3); phenylalanine (−2.5); tryptophan (−3.4).


“Endogenous” with reference to a polynucleotide or protein refers to a polynucleotide or protein that occurs naturally in the host cell.


As used herein, Enzyme Classification (EC) Numbers (EC numbers) are established by the Nomenclature Committee of the International Union of Biochemistry and Molecular Biology (IUBMB), description of which is available on the IUBMB Enzyme Nomenclature website on the World Wide Web. EC numbers classify enzymes according to the reaction catalyzed. For example, enzymes classified under EC 3.1.2.- or EC 3.1.2.14 are acyl-ACP hydrolases that cleave the thioester bonds from acyl-carrier proteins in the presence of water. On some embodiments, the enzyme is an acyl-ACP TE classified under EC 3.1.2.-. In some embodiments, the enzyme is an acyl-ACP TE classified under EC 3.1.2.14. An exemplary sequence classified under EC 3.1.2.14 is the sequence set forth under UniProt ID No. B1MVT0 from Leuconostoc citreum, hereby incorporated in its entirety by this reference. In other embodiments, the enzyme is an acyl-ACP TE classified under EC 3.1.1.1, EC 3.1.1.2, or EC 3.1.1.5. In specific embodiments, the acyl-ACP TE catalyzes the conversion of pimeloyl-ACP to pimelic acid.


As used herein, “expression” refers to the process by which a polypeptide is produced based on the nucleic acid sequence of a gene. The process includes both transcription and translation.


An “expression vector” as used herein means a DNA construct comprising a DNA sequence which is operably linked to a suitable control sequence capable of effecting expression of the DNA in a suitable host. Such control sequences may include a promoter to effect transcription, an optional operator sequence to control transcription, a sequence encoding suitable ribosome binding sites on the mRNA, enhancers and sequences which control termination of transcription and translation.


Examples of routinely used “expression systems” include recombinant baculovirus, lentivirus, protozoa (e.g., eukaryotic parasite Leishmania tarentolae), microbial expression systems, including yeast-based (e.g. Pichia pastoris, Saccharomyces cerevisiae, Yarrowia lipolytica, Hansenula polymorpha, Aspergillus and Trichoderma Fungi) and bacterial-based (e.g. E. coli, Pseudomonas fluorescens, Lactobacillus, Lactococcus, Bacillus megaterium, Bacillus subtilis, Brevibacillus, Corynebacterium glutamicum), Chinese hamster ovary (CHO) cells, CHOK1SVNSO (Lonza, Basel, Switzerland), baby hamster kidney (BHK) cells, PerC.6 or Per.C6 cells (e.g. Percivia, Cambridge, Mass.; Crucell, Leiden, Netherlands), different cell lines of HEK 293, Expi293F™ cells (Life Technologies, Carlsbad, Calif.), GenScript's YeastHIGH™ Technology (GenScript, Piscataway, N.J.), human neuronal precursor cell line AGE1.HN (Probiogen (Berlin Germany) and other mammalian cells, plants (e.g., corn, alfalfa, and tobacco), insect cells, avian eggs, algae, and transgenic animals (e.g., mice, rats, goats, sheep, pigs, cows). The advantages and disadvantages of these various systems have been reviewed in the literature and are known to one of ordinary skill in the art.


A “gene” refers to a DNA segment that is involved in producing a polypeptide and includes regions preceding and following the coding regions as well as intervening sequences (introns) between individual coding segments (exons).


“Host strain” or “host cell” means a suitable host for an expression vector or DNA construct comprising a polynucleotide encoding a polypeptide according to the disclosure. Specifically, host strains may be bacterial cells, mammalian cells, insect cells, and other cloning or “expression systems.” In an embodiment of the disclosure, “host cell” means both the cells and protoplasts created from the cells of a microbial strain. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.


“Heterologous” with reference to a polynucleotide or protein refers to a polynucleotide or protein/polypeptide that does not naturally occur in a host cell. In some embodiments, the protein is a commercially important industrial protein. It is intended that the term encompass proteins that are encoded by naturally occurring genes, mutated genes, and/or synthetic genes.


The terms “identical” or percent “identity,” in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same (e.g., 60% identity, optionally 65%, 70%, 75%, 80%, 85%, 90%, or 95% identity over a specified region), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Sequences are “substantially identical” to each other if they are at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, or at least 55% identical. Optionally, the identity exists over a region that is at least about 50 nucleotides in length, or more typically over a region that is 100 to 500 or 1000 or more nucleotides in length.


As used herein, “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the sequence in the comparison window can comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.


For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters are commonly used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities or similarities for the test sequences relative to the reference sequence, based on the program parameters.


A “comparison window,” as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, for example, by the local homology algorithm of Smith and Waterman (Adv. Appl. Math. 2:482, 1970), by the homology alignment algorithm of Needleman and Wunsch (J. Mol. Biol. 48:443, 1970), by the search for similarity method of Pearson and Lipman (Proc. Natl. Acad. Sci. USA 85:2444, 1988), by computerized implementations of these algorithms (e.g., GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by manual alignment and visual inspection (see, e.g., Ausubel et al., Current Protocols in Molecular Biology (1995 supplement)).


When percentage of sequence identity is used in reference to proteins it is recognized that residue positions which are not identical often differ by conservative amino acid substitutions, where amino acid residues are substituted for other amino acid residues with similar chemical properties (e.g., charge or hydrophobicity) and therefore do not change the functional properties of the molecule. Where sequences differ in conservative substitutions, the percent sequence identity may be adjusted upwards to correct for the conservative nature of the substitution and this process results in “sequence homology” of, e.g., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%. Means for making this adjustment are well known to those of skill in the art. Typically this involves scoring a conservative substitution as a partial rather than a full mismatch, thereby increasing the percentage sequence identity. Thus, for example, where an identical amino acid is given a score of 1 and a non-conservative substitution is given a score of zero, a conservative substitution is given a score between zero and 1. The scoring of conservative substitutions is calculated, e.g., according to the algorithm of Meyers and Miller, (1988) Computer Applic. Biol. Sci. 4:11-17, e.g., as implemented in the program PC/GENE (Intelligenetics, Mountain View, Calif., USA).


“Introduced” in the context of inserting a nucleic acid sequence into a cell, includes “transfection”, or “transformation” or “transduction” and includes reference to the incorporation of a nucleic acid sequence into a eukaryotic or prokaryotic cell wherein the nucleic acid sequence may be incorporated into the genome of the cell (e.g., chromosome, plasmid, plastid, or mitochondrial DNA), converted into an autonomous replicon, or transiently expressed (e.g., transfected mRNA). The term can also refer to the translocation of the nucleic acid sequence from outside a cell to inside a cell. In some cases, introducing refers to translocation of a nucleic acid from outside the cell to inside the nucleus of the cell.


With regard to enzymatic activity, “Kcat (s−1)” is the overall catalytic rate of the enzyme, or the maximum number of enzymatic reactions catalyzed per second. This constant is also referred to as the “turnover number” of the enzyme or the number of substrate molecules each enzyme site converts to product per unit time. “Km” is the substrate concentration required for the enzymatic reaction to occur at one-half Vmax, or one-half its maximal rate.


As used herein, a metabolically engineered microorganism is an organism produced via the introduction of genetic material into a host or parental microorganism of choice thereby modifying or altering the cellular physiology and biochemistry of the microorganism.


The term “mutant,” refers to both polypeptides and nucleic acids. The term “mutant” may be used interchangeably with the term “variant” or “synthetic”. Mutants or variants include alterations, insertions, substitutions, transversions, truncations, and/or inversions at one or more locations in the amino acid or nucleotide sequence, respectively, of a parent sequence. In the context of the synthetic acyl-ACP TEs of the present disclosure, a mutant acyl-ACP TE means a polypeptide, typically recombinant, that comprises one or more amino acid modifications, for example, one or more substitutions, relative to a corresponding, acyl-ACP TE, for example, a wild-type Acyl-ACP TE. In some embodiments, the mutant acyl-ACP TEs of the present disclosure are non-naturally occurring.


As used herein, “nucleotide sequence” or “nucleic acid sequence” refers to an oligonucleotide sequence or polynucleotide sequence and variants, homologues, fragments and derivatives thereof. The nucleotide sequence may be of genomic, synthetic or recombinant origin and may be double-stranded or single-stranded, whether representing the sense or anti-sense strand. As used herein, the term “nucleotide sequence” includes genomic DNA, cDNA, synthetic DNA, and RNA.


The term “nucleic acid” encompasses DNA, cDNA, RNA, heteroduplexes, and synthetic molecules capable of encoding a polypeptide. RNA includes mRNA, RNA, RNAi, siRNA, cRNA and autocatalytic RNA. Nucleic acids may be single stranded or double stranded, and may be chemical modifications. The terms “nucleic acid” and “polynucleotide” are used interchangeably. Because the genetic code is degenerate, more than one codon may be used to encode a particular amino acid, and the present compositions and methods encompass nucleotide sequences which encode a particular amino acid sequence. A nucleic acid comprises a nucleotide sequence which typically includes nucleotides that comprise an A, G, C, T or U base. However, nucleotide sequences may include other bases such as, without limitation inosine, methylycytosine, methylinosine, methyladenosine and/or thiouridine, although without limitation thereto. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated. A nucleic acid can be or can include, e.g., a chromosome or chromosomal segment, a vector (e.g., an expression vector), an expression cassette, a naked DNA or RNA polymer, or the product of a polymerase chain reaction (PCR), an oligonucleotide, a probe, and a primer. Unless otherwise indicated, a particular nucleic acid sequence optionally comprises or encodes complementary sequences, in addition to any sequence explicitly indicated.


The term “operably linked” and its variants refer to chemical fusion or bonding or association of sufficient stability to withstand conditions encountered in the nucleotide incorporation methods utilized, between a combination of different compounds; molecules or other entities such as, but not limited to: between a protein and a reporter moiety (e.g., fluorescent dye or nanoparticle); between a nucleotide and a reporter moiety (e.g., fluorescent dye); or between a promoter and a coding sequence, if it controls the transcription of the sequence.


A “promoter” is a regulatory sequence that is involved in binding RNA polymerase to initiate transcription of a gene. The promoter may be an inducible promoter or a constitutive promoter. An exemplary promoter used herein is a T7 promoter, which is an inducible promoter.


A “periplasmic tag” or “periplasmic leader sequence” is a sequence of amino acids which, when attached to/present at the N-terminus of a protein/peptide, directs the protein/peptide to the bacterial periplasm, where the sequence is often removed by a signal peptidase. Protein/peptide secretion into the periplasm can increase the stability of recombinantly-expressed proteins/peptides. An example of a periplasmic tag is a PelB leader sequence derived from an Erwinia carotovora pectate lyase gene, disclosed herein as SEQ ID NO: 2 (MKYLLPTAAAGLLLLAAQPAMAMG).


“Polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. As used herein, the terms encompass amino acid chains of any length, including full-length proteins, wherein the amino acid residues are linked by covalent peptide bonds.


“Recombinant” when used in reference to a cell, nucleic acid, protein or vector indicates that the cell, nucleic acid, protein or vector has been modified by the introduction of a “heterologous nucleic acid” or protein, or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all. A recombinant nucleic acid can be originally formed in vitro, in general, by the manipulation of a nucleic acid by endonucleases, in a form not normally found in nature. Thus an isolated, mutant acyl-ACP TE nucleic acid, in a linear form, or an expression vector formed in vitro by ligating DNA molecules that are not normally joined, are both considered recombinant for the purposes of this disclosure. It is understood that once a recombinant nucleic acid is made and reintroduced into a host cell, it will replicate non-recombinantly, i.e., using the in vivo cellular machinery of the host cell rather than in vitro manipulations; however, such nucleic acids, once produced recombinantly, although subsequently replicated non-recombinantly, are still considered recombinant for the purposes of the disclosure. A recombinant protein can be made using recombinant techniques, i.e., through the expression of a recombinant nucleic acid as depicted above. A recombinant protein is typically distinguished from a naturally occurring protein by at least one or more characteristics.


A “signal sequence” or “signal peptide” means a sequence of amino acids bound to the N-terminal portion of a protein, which facilitates the secretion of the mature form of the protein outside the cell. The definition of a signal sequence is a functional one. The mature form of the extracellular protein lacks the signal sequence which is cleaved off during the secretion process.


“Selective marker” refers to a gene capable of expression in a host that allows for ease of selection of those hosts containing an introduced nucleic acid or vector. Examples of selectable markers include but are not limited to antimicrobials (e.g., hygromycin, bleomycin, or chloramphenicol) and/or genes that confer a metabolic advantage, such as a nutritional advantage on the host cell.


“Substrate” refers to a molecule that an enzyme binds to and converts to a product. Pimeloyl-ACP is an example of a substrate that is converted to pimelic acid by a polypeptide described herein.


“Under transcriptional control” is a term well understood in the art that indicates that transcription of a polynucleotide sequence, usually a DNA sequence, depends on its being operably linked to an element which contributes to the initiation of, or promotes transcription.


“Under translational control” is a term well understood in the art that indicates a regulatory process that occurs after mRNA has been formed.


As used herein, “transformed cell” includes cells that have been transformed or transduced by use of recombinant DNA techniques. Transformation typically occurs by insertion of one or more nucleotide sequences into a cell. The inserted nucleotide sequence may be a “heterologous nucleotide sequence,” i.e., is a sequence that is not natural to the cell that is to be transformed, such as a fusion protein.


As used herein, “transformed”, “stably transformed”, “transduced,” and “transgenic” used in reference to a cell means the cell has a non-native (e.g., heterologous) nucleic acid sequence integrated into its genome or as an episomal plasmid that is maintained through multiple generations.


The term “vector”, as used herein, refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. 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) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”). In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, “plasmid” and “vector” may be used interchangeably as the plasmid is the most commonly used form of vector. However, the claimed embodiments are intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions. Vectors also include cloning vectors, shuttle vectors, plasmids, phage particles, cassettes and the like.


The term “wild-type,” in the context of a nucleotide sequence or polypeptide sequence, refers to the major or most common allele of the sequence as it occurs in nature. A wild-type sequence can also be referred to as a naturally occurring typical or normal sequence without modifications.


Synthetic Polypeptides

It has been discovered that altering amino acid residues at specific positions in a polypeptide having an acyl-ACP thioesterase activity could improve enzymatic activity and/or substrate specificity, for example substrate specificity for pimeloyl-ACP.


The present disclosure provides polypeptides having an acyl-acyl carrier protein (ACP) thioesterase (TE) activity. In some embodiments, the polypeptide having an acyl-ACP TE activity is a polypeptide comprising one or more amino acid substitution(s) relative to a wild-type acyl-ACP TE, wherein the one or more amino acid substitution(s) are at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1, or a functional fragment thereof.


Also provided are polypeptides comprising, consisting essentially of, or consisting of a mutant acyl-ACP TE or a functional fragment thereof. In some embodiments, the mutant acyl-ACP TE comprises one or more amino acid substitution(s) relative to a wild-type acyl-ACP TE, wherein the one or more amino acid substitution(s) are at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1. SEQ ID NO: 1 is the polypeptide sequence of a wild-type acyl-ACP TE from Eggerthella sp. CAG:1427 (Uniprot ID No. R5FQ35). In some embodiments, the enzyme is an acyl-ACP TE classified under EC 3.1.2.-. In some embodiments, the wild-type acyl-ACP TE is classified under EC 3.1.2.14. In other embodiments, the wild-type acyl-ACP TE is classified under EC 3.1.1.1, 3.1.1.2, or 3.1.1.5. Examples of wild-type acyl-ACP TEs include, but are not limited to, the wild-type acyl-ACP TEs set forth in Table 1.












TABLE 1





Uniprot


SEQ ID


ID No
Organism
Amino acid sequence
NO:


















R5FQ35

Eggerthella

MTIGYEQTYQLRTGDFDRYAHLLPSSILDVFQDVAGVNAEQVPGMTWKELTDAG
1



sp.
LFWVVTRIKYEVIETPHLHEQVIARTWPLAPTRLGFQREYTMRKLDGTPLVKCSS




CAG: 1427
DWILMDYKTRSFASARDFYNGPQDFSEEKVFEKKLRKIKTFEPEDTGETFQVHFV





DIDINGHVNNSKYPNFVMNSLDLGEDETIKTFQIDYRHELRAGSTVRIHSKRDGN





VITSMGISTEGDTAGECMFATQIELA (SEQ ID NO: 1)






Q07792

Vibrio

MIRLLSLVLFFCLSAASQASEKLLVLGDSLSAGYQMPIEKSWPSLLPDALLEHGQD
3




mimicus

VTVINGSISGDTTGNGLARLPQLLDQHTPDLVLIELGANDGLRGFPPKVITSNLSK





MISLIKDSGANVVMMQIRVPPNYGKRYSDMFYDIYPKLAEHQQVQLMPFFLEHVI





TKPEWMMDDGLHPKPEAQPWIAEFVAQELVKHL






A0A0F7J

Sedimenticola

MKKLFLLLIFAMSNVPAAEPVILVLGDSLSAAYGIERSRGWVSLLQSRLQQAGYP
4


XA5

thiotaurini

HRVVNASISGDTTAGGLARLPRALEQFQPDILIIELGGNDGLRGLGNRQTRDHLD





QMITLARASHSRPLLLGMMLPPNFGKAFTEKFLQIYRDLAEQRNVPLVPFFLAGV





ADRPEWMQSDGIHPTAEGQPLMLEHVWEQLQPMLEETSVHSDKSLK






K5D7V3

Bacteroides

MSESNKIGTYKFVAEPFHVDFNGRLTMGVLGNHLLNCAGFHASDRGFGIASLNE
5




finegoldii

DNYTWVLSRLAIELDEMPYQYEDFSVQTWVENVYRLFTDRNFAIMNKEGKKIGY




CL09T03
ARSVWAMISLNTRKPADLLALHGGSIVDYICDEPCPIEKPSRIKVTNTQPLATLTA




C10
KYSDIDINGHVNSIRYIEHILDLFPIDLYKTKRIRRFEMAYVAESYFGDELIFFCDE





ANENEFHVEVKKNGSEVVCRSKVIFE






A0A0M9

Pseudo-

MSASILRVFIVLLLVTKPLFAWADNQILVLGDSLSAAYGLKQHQGWVQLLQDTY
6


UHQ1

altromonas

EQQNTPTTLINASISGETTGGALRRLDAILKEHTPTHVLIELGGNDGLRGFPVTKM




sp.
KANLHALIDKSREAGAEVALMQIRIPPNYGRRYTELFEQSFVDVAKEKQVTLMPF




SW0106-04
FVEQVATNGELMQNDNIHPNAEAQPILRDIMKTQISNWLTK






A0A0F9W
marine
MTVKALTTRLPAPGPQVRYFSALRSAVLCVFLLAAGFCASTSAESDDGVLLVFGD
7


7B7
sediment
SLSAAYRMDERDGWVALLQQQLREESTALQVVNGSVSGETTAGGLARLPAMLD




metagenome
AHQPDIVMLELGGNDGLRGLPVTSIRQNLERMIQMSQQAGARVILAGIQIPPNYGP





RYTAPFYAQYQELADEYGLVLIPFLLEGIADNPALMQDDGIHPTAAAQPMIVDTV





WPVLQGVLTATDRP






A0A0C3E

Vibrio

MIRLLSLFLFFSLSTLAHANEKLLVLGDSLSAGYQMPIEKSWPSLLSNALLEHDQD
8


BX5

mytili

VTVINGSISGDTTGNGLARLPQLLDQHTPDFVILELGANDGLRGFPPKLITANLSK





MITMIKNSGAKVFMMQIRVPPNYGKRYSDMFYDIYPKLAEHQQVTLLPFFLEHV





VTKPEWMMDDGLHPKPEAQPWIAEFVAQELIKHL






A6D1N2

Vibrio

MIRQLSYFVLIVISLASFHAKAATLLILGDSLSAGYNMRAEQSWPTMLSDELSSGD
9




shilonii

EPMKVINGSVSGDTTSNGLAKLPGLLKQHSPDYVLIELGANDGLRGFQPSIIKNNL




AK1
ASLIEMSQQAGSKVLLMQIRIPPNYGKRYASMFEGIYPALAQESGVPLLPFFLEQVI





IKPEWMMEDGLHPKPEAQPFIAQFVAESMQPHL






A0A0B7D

Pseudomonas

MRVWFLSAGLALMCMAQNAAAGTVLIVGDSISAGFGLDTRLGWVSLLEQRLEQ
10


FD2

fluorescens

EGFDDKVVNASISGDTSAGGQARLPALLAEHKPDLVILELGGNDGLRGMPPTQLQ





QNLAAMIDSSRQSGAKVLLLGMQLPPNYGKRYTDAFAEVYGKLADDKKIPLVPF





FLDGVGGHPDLMQADGLHPAAGAQGKLLENVWPTLKPLL






A0A0B4Y
Thalassospira
MTIQWDEIAKPALPANCGWRGTYRVRYSEIGDNGLAMLPALADYMQDAAGWG
11


4H4
xiamenensis
ARILKLAYDDTVDKGMAWVLARMVIHVRRYPGNGEDIIVETWPSGVARRVATR




M-5 =
DFRLIDSSGDVIAVAQSFWVMFDLLERRAASWPDWIEERLPKPPGPKLIEPPFRPPF




DSM
TTDPLPEIDSIKARPSDLDLYGHVNNVRLMQWVLGATGADSKPDFHPESIDIQFRT




17429
ECRVQERVTVRQKDGFAAITRDGDGVDLVRAHVVPKNRTALA






A4VL40

Pseudomonas

MRRWLKCGALALLCWTQGALAGTVLVVGDSISAAFGLETSQGWVHLLQERLVE
12




stutzeri

GDESWRVVNASISGDTTAGGLARLDPLLEEHTPEVVILELGGNDGLRGQSPVQLK




(strain
QNLADMIDRSREAGAEVLLLGMRMPPNLGQRYTRAFADAFDSLAQEKSVAYVPF




A1501)
LLEGVGGVAGMMQADGIHPTAEAQSQLLETVWPALEPLL






E1WY53

Halo-

MKSPVFKKKYQVSISNVNINKRLGLFGLLGYLQDIATLHAEIAGFGLDEMISSNSF
13




bacteriovorax

WVLVRQEIRMNKFPKWNDEIEIQTWSRTPQGMYAFREFEFFLNDEKVGSCSTAW





marinus

MILSGDTRRIKKPDFPIEKINPRLDSLLDYCAERIKVLDNFELVNEIKVRISDLDLN




(strain
MHVNNTKYTQWVLDSIPIELHKSAKLRNYQINFLKEAHLGDEIDIYRASSAKDES




ATCC
AHDTQFKGIRRSDQSTIFYVNIIADT




BAA-682/





DSM





15412/





SJ)





(Bacterio






vorax







marinus)








F7Z1I0

Bacillus

MANGSSLYKGQYHIELRDVDFTKKLKLSALFSLFQDIASLAAEDLGYGIETLEKK
14




coagulans

YKVAWILTRIRVDILRHPTWDENITIETWPLQPSKIDFDRDFLVKDHTGAVIIKAAS




(strain
KWVVMGLNDRKIKRTESINIHYPENRTERAIEGKFGKFKDFGGLEPAYQKVIGYS




2-6)
DIDFNGHLNNSKYVDYIMDCFLPDFHKRHPIHTIEINFNQEALPGDSITLYKDISKM





DEHELYVEGVNQTDHHTIFKSHITIH






E1SPF5

Ferrimonas

MVRRSLLILMLCLYQPGAWADQPSILILGDSLSASYGMSEAEGWVKKLQQNLPD
15




balearica

AQIINASVSGETSGGGLRRLPGLLQQHQPDWVFVELGGNDGLRGFQPTITENNIEQ




(strain
LITLSKASGAQVLLSEVMVPPNYGRRYAERFQQIYHGLAKEHEVELVPFFMTEIA




DSM
TDPNLMQADGIHPNREAQGRIAAFLLPWFEQAIAE




9799/





CCM





4581/





PAT)







A0A067A

Pseudomonas

MSAGLALMCMAQGAAAGTVLIVGDSISAAFGLDTREGWVALLEQRLKREGFDA
16


754
sp.
KVVNASISGDTSAGGQARLPALLAEHKPELVVLELGGNDGLRGQLPTQLQQNLA




P482
SMIDKSRAAGAKVLLLGMRLPPNYGKRYNEAFAKVYENLATEKQVPLVPFFLEG





VGGVPELMQADGIHPGQGAQARLLENAWPQLKPLL






F2JLT2

Cellulo-

MSRLKENYQVDFDVVDFTGKLSINGLCSYMQTVAAKHATKLGINFYKNGEKPTY
17




silyticum

YWILSRVKYEIDTYPRWEDLVSLETYPGGYEKLFAVRLFDLTDEKGELIGRITGDY





lentocellum

LLMDAEKGRPVRIKGATGPLSVLDFPYEGRKIDKIEVPEVVLREQIRKAYYSELDL




(strain
NGHMNNAHYIRWTVDMLPLEVLKENEIVSLQINYNASITYGVETKLIIGKNEAGN




ATCC
YLVAGNSLDDSVNYFTSEIILRKNK




49066/





DSM





5427 /





NCIMB





11756/





RHM5)





(Clostridium






lentocellum)








L1M6X0

Pseudomonas

MRVWLLSAGLALLCMAQNAAAGTVLIVGDSISAAFGLDTRQGWVALLEKRLKD
18




putida

EGFDDKVINASISGDTSAGGQARLPALLSAHKPSLVVLELGGNDGLRGQLPAQLQ




CSV86
QNLASMIDKSRAAGAKVLLLGMQLPPNYGARYTKAFAQVYSDLAVQKNVPLVP





FFLEGVGGHPELMQADGIHPAQGAQGRLLENAWPAIKPLL






L8M832

Pseudomonas

MRAWLLGGCLSLLLLAQEAMAGTVLVVGDSISAALGLESSQGWVSLLEKRLVEK
19




pseudo-

GYDKQVVNASISGDTSAGGLSRLPALLAEHKPELVIIELGGNDGLRGQPPAQLQQ





alcaligenes

NLAGMIDSSRAQGAAVLLLGMRLPPNYGARYTSAFAKVYSDLAEQKQVPLVPFF




KF707 =
LEGVGGVPELMQPDGIHPQANAQPRLLENVWPTLEPLL




NBRC





110670







G8Q1V9

Pseudomonas

MRVWFLSAGLALMCMAQNAAAGTVLIVGDSISAAFGLDTRQGWVSLLEQRLKA
20




fluorescens

EGFDDKVVNASISGDTSAGGQARLPALLAAHKPDLVILELGGNDGLRGQPPTQLQ




F113
QNLAAMIDSARASGAKVLLLGMQLPPNYGRRYTEAFARVYSTVAEEKKVPLVPF





FLKDVGGIPTMMQGDGLHPSVAAQGQLLENVWPTLKPLL






A0A075P

Alteromonas

MFYSLRNIAFVFLLLIPTFAQSDQHDSDEKEPNAKLLILGDSLSAAYGLRQEEGWV
21


0V4

australica

SLLQNTWRDENIPIDIVNAAVSGETTDGGLARLPRLLTQHQPSHVLIELGGNDGLQ





GHNVKKIRSNLVALVKIAQSADAKVFLQDMQIPTNYGKRYTNMFGESFDRVGEE





LNVPVIPFFLQNIALDTSLMQRDGIHPNAEAQALIAEFMHRQLMPLFDN






K1LL76

Cecembia

MKESSNKPFQFQKTFEILSFQIDPSGKLRWAALADLLQEVAWKHADSREFGQVLF
22




lonarensis

DKGFMWVLSRFDIQVHAMPSWGETIHIETAGRGINKLFALREFRVTDSSGTVLAT




LW9
AMSAWLLLDIKTKRPQRPSLVLPSELFETEPSDYAPPEKISVPEKGHTGKTFHVNH





SDLDMNNHVNNVSYIRWIEDFCLEQGFTFDKISINYLNEALLGENIEILFSLDAQK





MLVSGRSGERDVFTSWVEKLNG






A0A0F7M

Spongiibacter

MTHFIQFLPGRISPRRALALLLVLLLQGALPRIVLADTILLLGDSLSAAYKIPVESS
23


706
sp.
WPALLQDSIDEEHSLKNASVSGETTAGGLARLPALLAKNNTDILIIELGGNDGLRG




IMCC21906
YPLVRIRENIEKMIKLGQKKGAKVVLLGMHIPPNYGRRYADGFHNIYLSLAEENN





TALLPFLLDGVAKQPRLMQGDGIHPTAEAQPIILQNVLSVLRPLLDSTN






G7LRH3

Brenneria

MAADTLLILGDSLSAGYRMPAASAWPALLDQKWQTRPDGVKVVNASISGDTAG
24



sp.
QGLARLPALLKQHQPRWVLIELGGNDGLRGFPPNNIEQDLSKIITLVEQAQAQPLL




EniD312
MQIRLPTNYGRRYNESFSDVYPRLAKQFSIPLVPFFMEQVYLKPEWILEDGIHPAR





DAQPFIADWMAQQLESLYPIDFELQEGGN






H7FRY9

Flavo-

MPIASNFTSVLSKDWEINFTQCMPNGYLKYTDLCNILQLTAAAHSDMGGISFSDM
25




bacterium

QEFNQAWVLSRMRVEIAALPKWRDIVTVKTWINTLENSRSVRALEMYVNGEKIV





frigoris

GSETFWAVFNTERRRPEGLALPYEHFELYPELKATKESFSKININSEKEDVFEKSIY




PS1
LSDLDIVNHVNNVKYLEWCLDHLEVDLILSQKIRSFEMNFLKELSLYDKVIIHENY





SEDSILFSITKENKNCYALQLNL






H2FZ27

Oceanimonas

MLRILVLLLCLVSPTVLADTLLILGDSLSAGYRMRSDQAWPHLLAEQWRREGRQ
26



sp.
VTVINASVSGDTTQGGLQRLPPLLQRHQPSLVLLELGGNDGLRGLPPGLIERNLER




(strain
LIALAGDAGARVILTDIQLPPNYGRRYLQQFEQVFSRLASQHQLPLLPFFVAPLMG




GK1)
EQGMMMDDGIHPTVKAQPLIARQVGEFLTPYLTP






12BBI6

Shimwellia

MMHFKNMLRWHLPFLLLVLLMCRTAAADTLLVLGDSLSAGYRMPAEEAWPAL
27




blattae

LDKQWEKRQIRVINASISGDTAAQGLARLPTLLAEHKPRWVVIELGGNDGLRGFP




(strain
PAGIAATLSQIISQVKAARARPILVQIHLPANYGRRYNESFGAIYATLAASNDIPLL




ATCC
PFFMEEVYLKPQWMQDDGIHPNASAQPFIADWMAQHLNPLVNHDS




29907/





DSM





4481/





JCM





1650/





NBRC





105725/





CDC





9005-74)





(Escherichia






blattae)








M1WJV0

Pseudode

MVSLHPFPSQLHKIVMTTNIDLAFEHLYPVQSYEPRMDGRIAIPSVCNYLQDIASR
28




sulfovibrio

HADTLGFGYHDLEKCGHFWMLARLHVTMERLPRFGESCRIETWPSGNERLVALR





piezophilus

DFLLHDEKGLIGKATTSWVTVNTTTHKPDNPETVLNRRFIPKRDRATIFPTKAIKR




(strain
LKGGEHDIRLVARRSDMDINNHVNNVHYVEFCLEAIPRSWEEKNRCLGIDIQFRS




DSM
ESHAGDEYVSACAPADPDGPHATFLHSLTRLSDGREVVRMRSWWIQA




21447/





JCm





15486/





CITLV30)





(Desulfo-






vibrio







piezophilus)








J3H5M3

Pseudomonas

MRVWFLSAGLALMCMAQNAAAGTVLIVGDSISAGFGLDTREGWVSLLEQRLKR
29



sp.
EGFDDKVVNASISGDTSAGGRARLPALLAEHKPELVILELGGNDGLRGMLPTQLQ




GM67
QNLAAMINSSKASGAKVLLLGMQLPPNYGARYNKVFAEAFSNVAAEKKIPLVPF





FLEGVGGHPELMQSDGLHPAAGAQDKLLENVWPTLKPLL






E6W9H0

Pantoea

MLSLTDGLSKMMNFNNVFRWHYSFLLLLLLLVSRLAAADTLLVLGDSLSAGYR
30



sp. (strain
MSANVAWPYLLDKEWQQQPKVINASISGDTAGQGLARLPALLKQHQPRWVLIEL




At-9b)
GGNDGLRGFPPQNIAQDLSKIIDDVKAANAQPLLMQIRLPANYGRRYTQAFSAIY





PQLAQQYNIPLVPFFMEQVYLKPEWMQQDGIHPNPDAQPFIASVMAKELAPLVK





HD






T0CHD7

Bacterio-

MKDKVFKKTYQVSINSVNINKKLGLFGILGYLQDIATVHAEVMGFGLEDMIRDQS
31




vorax sp.

FWVLVRQKLRMTKFPVWNESVEIQTWSRPPEGMYAFREFEIFLDGEKIGDCSTV




BAL6)X
WMILDGVIRKVKKPDFSMERINPRTDYHLDYIAHKVEVRDNFEKVNTITVRNSD





LDLNMHVNNTKYSQWILDSIPIELHKTATLNEFEINFMAETHLGDEIDIYRARNVE





GEFKHDITYKGVRHSDGKTCFLAKLLAD






A0A0C4

Azotobacter

MLGGALASLLFWAQGALAGTVLVVGDSISAALGVETSQGWVALLERRLVDQGL
32


WQF1

chroococcum

THRVVNASISGDTSAGGLARLPTLLATHRPELVIIELGGNDGLRGQPPQQLQQNLA




NCIMB
AMIDSSHSSGAQVVLLGMQLPPNYGPRYNQAFSRVYATLAEEKQVPLVPFFLDG




8003
VGGVPGMMQADGIHPTAKAQPKMLDNLWPTLEPLL






D2TLW8
Citrobacter
MVNFNNVFRWHLPFLFLILFTCRSVAADTLLILGDSLSAGYRMSASAAWPALLND
33



rodentium
KWHSKTTIVNASISGDTSGQALARLPALLKQHQPRWVLVELGGNDGLRGFSPAQ




(strain
TEQTLRTILQNVKAASAEPLLMQIRLPANYGRRYNETFSAIYPKLASEFDIPLLPFF




ICC168)
MEEVYLKPQWMQDDGIHPNRDAQPFIADWMAKQLSPLVKYES




(Citrobacter






freundii







biotype






4280)







K5YG62

Pseudomonas

MRSWLKGGVLLLVMWAQGALAGTVLVVGDSISAAFGLETSQGWVHLLQQRLA
34



sp.
EQARPRSVVNASISGDTSAGGLARLPTLLAEHRPEVVILELGGNDGLRGQSPAQL




Chol1
KQNLAAMIEQSQQADAKVLLLGMRLPPNYGRRYTEAFARVYHELADERDVALV





PFVLEGVAGEPGMMQGDGVHPTASAQAQLLENVWPALAPLLAPQR






E1RAP4

Sediminis

MKQVSRYTTEHTVMYSETDARGVLSLPSFFALFQEAALLHAEELGFGETYSKQE
35




pirochaeta

NLMWVLSRLLLEIDAFPKHRDRIRLSTWPKQPQGPFAIRDYILESEEGTVCARATS





smaragdinae

SWLLLKLDTMRPIRPQTIFANLSMEGIGLAVEGTAPKISEIDNDSKQEMEVTARYS




(strain
DLDQNNHVNNTRYVRWFLDCYTPEEITTSGNLHFAINYLQAASYSDKLLLRRYD




DSM
TESDSSVYGYLEDGTPSFSARIERKSD




11293/





JCM





15392/





SEBR





4228)





(Spirochaeta






smaragdinae)








A3DJY9

Clostridium

MQKKRFSKKYEVHYYEINSMQEATLLSLLNYMEDCAISHSTSAGYGVNELLAAD
36




thermocellum

AGWVLYRWLIKIDRLPKLGETITVQTWASSFERFYGNREFIVLDGRDNPIVKASSV




(strain
WIYFNIKKRKPMRIPLEMGDAYGIDETRALEEPFTDFDFDFEPKVIEEFTVKRSDID




ATCC
TNSHVNNKKYVDWIMETVPQQIYDNYKVTSLQIIYKKESSLGSGIKAGCVIDEQN




27405/
TDNPRLLHKIWDKNTGLELVSAETIWQKIQS




DSM





1237/





NBRC





103400/





NCIMB





10682/





NRRL B-





4536/





VPI





7372)





(Rumini-






clostridium







thermocellum)








A0A0F2P

Gamma-

MLLIILLSCYFGTFSSASLANTILVLGDSISAAYRIPVESGWVSLLQERLDEHRPGH
37


667

proteobacteria

YTVVNASISGDTTAGALNRLPPLLEKHRPDQVIVELGGNDGLRGLPLKGMRDNL





bacterium

QAIIDLSRQQGADVVLVSIDLPTSYGSHFNQRFTQVYDELEESNKLPRVSLGFKLL




BRH_c0
NDRNLIQEDGIHPTEEAQPLLLDVVWPLIAPGVEREG






A0A0B3

Terri-

MERVFTKEYEVTYRDTDARGECFLTSYMNFMADCGLSQDEKYGFVIADMVKEN
38


WUQ1

sporobacter

HTWMLVDYEITIYKYVKYKEKLRAITYIEGMNKFYAVRYFKIYNDKDDLILEGKT





othiniensis

LVILVDSKKRRPLSIPDEHYKAYGVEEKTPTIGRNKLKLSKCKNVDYKKEFNVRY





SDIDLNLHVGNVTYLGWILETIPFEIMTDYKIYSVKIKYQKELTYGDKVSVKTEME





YNDNNISAYHEIINESEEVVALLETHWNEI






A3D613

Shewanella

MAKTLGAFILLSVLTVTPAHAAKVLILGDSLGASYGMAEQSGWVALLQKNLPEH
39




baltica

QFTNGSVSGETTAGGLRRLPALLDSVAPDLVVVELGGNDGLRGFPPTQLENNLIQI




(strain
ITLAKDSGAKVLLTEIMVPPNYGPRYTQKFTQVYQDISKTQDIVLIPFFMQDIAPHP




OS155/
ELMQRDGIHPNEKAQAQIATWMQPWIEDALTQ




ATCC





BAA-





1091)







A3QFM1

Shewanella

MQAACLLVFFLIPQAHANPILILGDSLSASYGMEQDKGWVHLLQQQSPEVTIINGS
40




loihica

VSGETSAGGLRRLPALLDSTKAKRVFIELGGNDGLRGFSPKQLKNNLTKMILLAK




(strain
DSGAEVLLSEVMVPPNYGPRYAKQFTQVYQELSSEQGVTLVPFFMTEIAIHPELM




ATCC
QADGIHPNEQAQPQIVSFIRPWLIDTQQPSE




BAA-





1088/





PV-4)







A0A0C1Q

Lactobacillus

MAANEFSETHRVVYYEADDTGQLTLAMLINLFVLVSEDQNDALGLSTAFVQSHG
41


9A5

brevis

VGWVVTQYHLHIDELPRTGAQVTIKTRATAYNRYFAYREYWLLDDAGQVLAYG





EGIWVTMSYATRKITTIPAEVMAPYHSEEQTRLPRLPRPDHFDEAVNQTLKPYTV





RYFDIDGNGHVNNAHYFDWMLDVLPATFLRAHHPTDVKIRFENEVQYGHQVTS





ELSQAAALTTQHMIKVGDLTAVKATIQWDNR






A0A0K8Q

Bacteroidales

MVPLKHVHHLKAKSYHINRFGEVSTPFLFWYMQEIAWEHAHKLGFGFEHLKEDQ
42


XH4

bacterium

LFWVLSRLLVKIDHRPRWTDEFTLETWSRGTDGFFAYRDYRFLDQNGNEFIKATS




6E
SWLVLDLESRRIQRLSQFKNFPVYQESVLGNNAGKVDTPETLGDLSFFPVLFNEID





INQHFNTGRYLERINNSYSFDFHENHTLSELEVNFIKEGMADDSLAVCQQRLSEGE





HLCSVIRQRDGSELIKARLVWEQKKKI






A1WYS0

Halor-

MAGSSCFSLSLRRALCAAALALLLGPSAATAERPTILIFGDSLSTAYGFDRDEAWP
43




hodospira

VLLEARLDEADRPHRVANVSRSGETTSGGTRRLPDALEEHEPEIVLLQLGGNDGL





halophila

RGQPPERIRSNLQQMIEQARAVDSRVLLIGIRIPPNYGRTYTEQFAAIYPELADEQD




(strain
VPVIPFLLEGVWDRDGMMQDDGVHPTAKAQPEIAETVWETLREMLDGPS




DSM 244/





SL1)





(Ectothio-






rhodospira







halophila






(strain





DSM 244/





SL1))







P44886

Haemophdus

MSANFTDKNGRQSKGVLLLRTLAMPSDTNANGDIFGGWIMSQMDMGGAILAKEI
44




influenzae

AHGRVVTVAVESMNFIKPISVGDVVCCYGQCLKVGRSSIKIKVEVWVKKVASEPI




(strain
GERYCVTDAVFTFVAVDNNGRSRTIPRENNQELEKALALISEQPL




ATCC





51907/





DSM





11121/





KW20/





Rd)







D6E2B 1
[Eubacterium]
MAFSFDSRIRYSEVDSSCRLSLTGLTNYFQDCSVFHSQSHDVGIRFLADNHIAWVL
45




rectale

SSWQICINRLPLLNEQVKISTWAYGMKAFYGYRNFTLEDAGGSTLAYANSVWVL




DSM
VDTRTGRPVKVPQEFADTYGLEPQLEMECAKRKLHIPDDMEKKGEIEVPQFFIDS




17629
NHHMNNEKYVMLAQQLLPNDFEISELRVEYRKEAKLGDTVISYVKYTSKSVTVV





LADTDKKPYSVVEFLSKPVPGK






Q69MT1

Oryza

MAATRSTATAALALSRTLARRPAASSSSRRISLELSAPRGTNPFQSAAFSSTTTGDP
46




sativa

PPPTMDSPIKVVSHIGGSGGDGGGGAIDAGRSARKPLSLWPGMYHSPVTNALWE




subsp.
ARSSIFERMIDAGAAGKQQQQPPQTELLTKTPAGSRTSIVYKFATDDILREQYRDP





japonica

WNEVRIGKLLEDLDALAGTIAVKHCSDEDSTTRPLLLVTASVDKMELKKPICVDT




(Rice)
DLKIAGAVTYVGRSSIDIQIEVTQVDQDSDMQSDPIALTANFTFVARDSMTGKSAP





VNRLSPETEKEKQLFAEREARDKLRKRKREEQKGVFENGINKLHVEAERLNSLLA





EGRVFSDLPALADRDSILLKDTRLENSLICQPQQRNLHGRIFGGFLMHRAFELAFS





TAYAFVGQRPCFLEVDHVDFLKPVDVGDFLRFKSCVLYTQLDNAEQPLVNVEVV





AHVTRPELRKSEVSNTFHFTFTVCSDALKNGLKIRHVVPSTEEEARRILERMDAEG





LFD






A0A0K6I

Marinomonas

MMSKVCVVLLCFLSLLTNAHANTLLVFGDSLSAAYNLRQQQGWVSLLSQQLNR
47


SB1

fungiae

SHPDVNVVNASISGETTQGGLSRLPKLLETHQPKWVVLELGANDGLRGYPLNQM





KQNLAHMIDQSQQTGAKVLLVGNHLPSNYGRTYTTQFFNVYSELAKEYQLAYVP





FMLENVALDSTLMQNDGLHPNADGQPVVLENIAPTLLPLLNLP






A4Y830

Shewanella

MAKTLGAFILLSMLMATPVHAAKVLILGDSLGASYGMSEQLGWVAMLQKNLPE
48




putrefaciens

HQFINGSVSGETTAGGLRRLPALLDSVSPDLVVVELGGNDGLRGFPPTQLENNLIQ




(strain
IITLAQKSGAKVLLTEIMVPPNYGPRYTQKFTQVYQDISKTQNIELIPFFMQEIAPY




CN-32/
PDLMQRDGIHPNEKAQAKIAAWMQPWIEKALNQ




ATCC





BAA-453)







K1J0J2

Aeromonas

MVRILFALFVGLGSLLSSAQAQTLLVLGDSLSAGYQMQVEQSWPALLNQKWQEE
49




veronii

GSKHTLLNASISGETTQGALARLPALLKEHKPDWLLIELGGNDGLRGFAPTITRQN




AMC35
LASMIALAKEQQTRVVLTQIQLPRNYGARYLRQFEQIFPELAQANDLPLLPFFMD





DIALRPELMMNDGIHPTPAAQPQIRDKVARFMEPLLSQ






D8QRX8

Selaginella

DTLKQGRLVERVVYRQTFVVRSYEVGPDKTATLDTFLNLFQETALNHVLISGLAG
50




moellend

NGFGTTHEMIRNNLIWVVTRMQVQVERYPAWGNALEIDTWVGASGKNGMRRD





orffii

WLVRDYKTGSILARATSTWVMMHKDTRRLSKMPDLVRAEISPWFLSRTAFIPEES




(Spikemoss)
CSKIEKLDNSNTRYIRSNLTPRHSDLDMNQHVNNVKYLTWMMESLPQNILESHH





LVGITLEYRRECSKSDMVESLTHPERGGHLAINGAAAAAAAAAAAPPSQLDFIHL





LRMQTGGSEIVRARTSWKSRH






A0A085A

Trabulsiella

MDFKYIFRWHLPFLLLVLFAFRAAAADTVLVLGDSLSAGYRMAANAAWPALLN
51


ET1

guamensis

EKWQPQTRVINASISGDTAQQGLARLPALLKQHQPDWVLVELGGNDGLRGFQPQ




ATCC
QTEQTLRTILQDIKAANARPVLMQIRLPANYGRRYNEAFSAIYPKLAGEFSVPLLP




49490
FFMEEVYLKPQWMQDDGIHPNRDAQPFIADWMAARLAPLVNHDS






F0P329

Weeksella

MNPVDYYTEKFYIDYSRVYPNRKIKYPELANILQITAANHADFCGLGFDDLQHNK
52




virosa

QAWVMNRIRIEIDTLPELNDEVTIDTWLELLRVPKSIRNLEIKKEGKKLVGVSSLW




(strain
AVFNTERRRPEALKIDADHLKIFTDLHATSLENNKIETPESFQKVAEYQVKLSDLD




ATCC
VVNHVNNIQYLTWCLDTLSKEEVLERSIAVLEMNFLKELSYQKIIHIEQYRQEHEL




43766/
YLRIRDEQFIYFVARITYQ




DSM





16922/





JCM





21250/





NBRC





16016/





NCTC





11634/





CL345/78)







F3BKQ8

Pseudo

MTHSILRLVFILFLVIKPLSAAADNTILILGDSLSAAYGLQQEEGWVKLLQDKYDD
53




alteromonas

EQADIELINASISGETSGGALRRLDALLEQYEPTHVLIELGANDGLRGFPVKKMQT





haloplanktis

NLTSLIQKSQAANAMTALMEIYIPPNYGPRYSKMFTDTFSRVSEETNAHLMNFFM




ANT/505
LDIASQSDLMQNDSLHPNKKAQPLIRDEMYDSIKKWLNKD






P0ADA2

Escherichia

MMNFNNVFRWHLPFLFLVLLTFRAAAADTLLILGDSLSAGYRMSASAAWPALLN
54




coli

DKWQSKTSVVNASISGDTSQQGLARLPALLKQHQPRWVLVELGGNDGLRGFQP




O6:H1
QQTEQTLRQILQDVKAANAEPLLMQIRLPANYGRRYNEAFSAIYPKLAKEFDVPL




(strain
LPFFMEEVYLKPQWMQDDGIHPNRDAQPFIADWMAKQLQPLVNHDS




CFT073/





ATCC





700928/





UPEC)







A0A0F4L

Lactobacillus

MGQQYQETKVIPFYSTNATSEINISALFNEMLLVSEHQLHAVGIDSQQMVAQGIG
55


189

mellis

WVVTKYHLEIKRLPRINEKVTIVTEANSYNKFFCYRTFTLYDSQGQQLLHLLSNW





VMMDIKKRSMIEVIPETMAKIGCEYSTDIWRFPRIQRFKYSQSPQIYRTRFFDIDVN





GHVNNSIYLDWMLDSLGKDFLMQHQLQTLDIKYDREVAYGQSVQSFVQLEDNLI





SHHQILTDNKVNAQAQMQWQQRQK






Q74KZ2

Lactobacillus

MEKIFKEEHQVSYGDCDETGKIQLPHLIEHFMQVSNDQLTAGGAGIHDLLKQNLG
56




johnsonii

WVVVEYHLDIDRLPEAGEKITVTTNGSGYNRFFEYRDFGIIDSTNKKIVGVKSQW




(strain
VILDLKNRKITEADDQMMQKFGNPYLKHMPRFKRLRPLKEYRSSKKYTVRYYDL




CNCM1-
DTNHHLTNSIYFDWMIDTLPREFLNSHTVKSIDISFKKEVQYGDQALAEVELDQD




12250/
TLTSYHLISNQGEASALAEINWKEN




La1/





NCC 533)







G7V8P3

Thermovirga

MYEHNFRISYSQAGALGRLKLTGAMNLCQDIADDHAERVGVSVADLLKQSKTW
57




lienii

VLHRFKMTIQTMPQRGDLVTIKTWYRPEKNLYSLRNFEMLDCNGKKLLSVQTSW




(strain
VVVDMNRGRPLRLDRVMPEAYDKNKDENLEVSFQELLLPEKVDVKKTIQVAVT




ATCC
DLDMNFHVNNVHYLRWALDTIPVEILKEYKPKGVEIAFKRPAFYGDSVISEVGID




BAA-
KNSCSILCRHHIYGEKDGQSMAVISTEWEKISREER




1197/





DSM





17291/





Cas60314)







C7ML86

Crypto-

MTISASLFLEKDYRLRTGDFDRYRRLHPTAVLDLFQDIGGLQAEMMGIGYDAMA
58




bacterium

AQDVFWAVVRTAYQVEHTPAEHEVVKVSTWPHSPSRYSFQRDYALRSTDGSLL





curtum

VRGTSEWVLMDMNTRSLTSVLDYYHGSLDFIDERMFAKKLRKIRDFTEEGSGLSI




(strain
TPRYSDVDQNGHVNNARYASFVLDALDPSAAGSIASFQIDFRHEVIEGTPLVVFTQ




ATCC
VEGKDIQAKGLDSNGEIKFACKITAQA




700683/





DSM





15641/





12-3)







F5YIQ3

Treponema

MDVWKESYPVGFTAVDESEGLTLAAAFDYFQEAARRHAEVLGVGQEPMVQAG
59




primitia

QGWVLSRISVLVERRPRQGELITVSTWPRGWEKLFALRDFDIRDESDKPIVRARSC




(strain
WLIVNIEKRRPLRPQATMEKLPLNEGRDALPGGGVGLAPLENLLKAGDRVAAYS




ATCC
DIDYNGHVNNARYVQWVQDIADPTALVQAKTLRLDINYLSEVKIHEPIELWTEPL




BAA-887/
PAESDAVYTLGVEGRRNGGAVFRAELRIKD




DSM





12427/





ZAS-2)







F7R2D3

Lactobacillus

MESKGFSEKHRVTYYETDMNGTVGLGRLVDLMMLSCNDQSDAVGLSSEKVNQ
60




ruminis

MGLGWIVTQNMIDIKRLPRRNETIYITTHAKSYNRYFCYRDFWIHDIRKQELAHM




SPM0211
HTVFALMDQNERKIVRIPENLIEPYHSEYATKIERLPLPKELERIDRQKEYEVRFW





DIDINQHVNNVHYFEWMLDALDLDFLIKYQPVSMNIEYKKEIRYGQKAVSQAQIT





MTGVETVTTFHEIKVNDELSCRAVCDWKLRKEEWIM






I7K130

Lactococcus

MLTYKKKYTVPYYETDANGNMKLPSLFNIALQLSGEQSHSLGISDDWLKETYNY
61




raffinolactis

AWVVVEYDVTIQRLPRFSEIITMSTFAKSYNKFFCYRDFVFYAENGDTLLTINSTF




4877
VLIDTTSRKVAHVEDDIVAPYQSEKISKIVRGHKSTALSDTPLEKSYHVRFNDIDQ





NGHVNNSKYFDWMTDVLGYDFLSSHVPSRIHLKYSKEVLYGATVTSRVDLVGV





QSFHEIVSEGKHAQAEMTWREK






D4YGM6

Aerococcus

MINIRHYQGGKRMGLLYQESVKVRHYHCNALGEMTLPAILDIMLIASNNQEATIP
62




viridans

EAKEGFRQEGWAWIITQNQIDINRLPRYDEDIIAETEATTYNKFFSKRHYALKTRD




ATCC
GLVLAQAETTFALIDLNQRSIVRIPEIVAEWYQVEKEERPSRRKRLNKEIAVESKL




11563 =
DRFEVKFLDIDLNNHVNNTIYLRWITNSLGMEWFEKYTPTSFTVAYEKEMYLHQ




CCUG
EGAVHSDISTVSEDLKSGDTFNSQHVIDSEDKAHCLTEITWQVK




311







U2CXE7

Clostridiales

MPGFVYEKEYEIHYYEIDYKRRALITSLVDFFGDIATVQSEQLGIGIEYLKENNLA
63




bacterium

WVLYKWNIDVVKYPLHGEKIIVKTCPYSMKKFYAYRTFEVLNSEGEVIATADSIW




oral
FLINIERRRPVRINEDVYRLYGLDYNDQNTLEIEDIKKPDKADLEKIFNVRYSDIDT




taxon 876
NQHVNNAKYIAWAIETVPMEVVLNYTIKNLKVIYEKETTYGEIVKVITEIIHNDNT




str.
VICIHKIIDKEEKELTLIKTTWEKNF




F0540







A0PXB0

Clostridium

MSGIITEKQYEIHYYEAHLKQQATITNIIDFFTDVSTFQSEELGVGINYMMENNMA
64




novyi

WILYKWDINVERYPRYREKILAVTEPYSIKKFYAYRKFYILDENRNIIASAKSTWL




(strain
LIDTKKRRPLRISKEMVKAFGLENKEETLEIENVHKLPEENTEINFKVRYSDIDTNG




NT)
HVNNEKYVAWMIESIPRDIILNYTLKNTKITYKKETMYGESIKVITGKIKEDEDKV





KFVHNILRENGELLTEGETLWEKNK






P44679

Haemophdus

MLDNGFSFPVRVYYEDTDAGGVVYHARYLHFFERARTEYLRTLNFTQQTLLEEQ
65




influenzae

QLAFVVKTLAIDYCVAAKLDDLLMVETEVSEVKGATILFEQRLMRNTLMLSKAT




(strain
VKVACVDLGKMKPVAFPKEVKAAFHHLK




ATCC





51907/





DSM





11121/





KW20/





Rd)







J0YTE5

Streptococcus

MGLTYQMKMKIPFDMADMNGHIKLPDVILLSLQVSGMQSINLGVSDKDVLEQY
66




infantis

NLVWIITDYDIDVVRLPQFDEEITIETEALTYNRLFCYRRFTIYDEDGQEIIRMVATF




SPAR10
VLMDRDSRKVHPVVPEIVAPYQSEFSKKLVRGPKYTELENAINKDYHVRFYDLD





MNGHVNNSKYLDWIFEVMGADFLTNHIPKKINLKYVKEVRPGGMITSSYELNQL





ESNHQVTSDGDINAQAKIIWQEINTD






I2F781

Mesotoga

MKPVITKEVYKVRYYELDCHWKASVSTLMDYFNDIVTLQTVEIGHGVDIMSKGE
67




prima

YAWLLLRWDVNVNRYPDYMENVVVQTIPYSMDRFYAYRRFEIFDRSDNLIVDA




MesG1.A
NSQWILIDQRKRRPIRIGDQFYALYGIDSDFHQPLSFPQVNDNDSYGEEITFRVRSS




g.4.2
DLDTNGHSNNVSYVRWIMETVPDEFAKRSLRRLTIEYKRESREGDEISVESVFEN





GDEFAEGKHKIISSGRVLSLARTEWK






R5BH39

Bacteroides

MGKYCEKDIITCYRSDSHHKMRSEAFLDFAQQLAVKGAQLLSFNDTALSQLGCI
68



sp.
WVLARMHVRFERDVAFDEKVDLSTWHKGQSGLYFLRDYQLCDRHGAAVNATS




CAG: 1060
SWIVMNAETRHISRDEKVLELLSVGPQSEDHAIKEASPKITVPKDCTLEVIGEHTV





RYSDVDYNNHANNVKYTVWALDHLPDNIAMTRRLKELSINFNKETLLGETVTLY





HCITPEGEHIVEGRSGDIQVFIEKLLFE






E3GJ26

Eubacterium

MGIIYEKKQKINGYECTYNYQLQPTAALNYFQQTSQEQSEQLGVGPEVLDEMGL
69




limosum

AWFLVKYKLQFHEYPKFNDEVMVETEMAFDKFAAHRRFAIKSLDGRMMVEGD




(strain
TEWMLQNRKENRLERLSNVPELDVYESGHENHFKLKRVAKVEEWTESKNFQVR




KIST612)
YLDIDFNSHVNHVKYLAWALETLPLEKVKAGEIETAKIIYKNQGFYGDMITVKSA





EIDENTYRMDIENQEGILLCQIEMTMRIRED






F4LT66

Tepidanaero-

MSGSNILEKEYRIHYYEVNAKGRVLIASLMRYFDDIATQQSRELGVGINYLKEHN
70




bacter

VAWMLYQWDIKINKYPRYGETIKVRTAPYSFRKFYAYRWFDVLNKDGETLVNA





acetatoxydans

NSVWLFVDTDKRRPLKIPDVMYEAYQVTSDEPLEIGEITELSSCDIEKKFQVRYSD




(strain
IDTNSHVNNVVYVTWALEALPYDIISNYELRNLKVTYKQETTYGMIIRSQAQVIKS




DSM
DDEVITRHSILSEEGKKLCLLEGRWIKNS




21804/





JCM





16047/





Re1)







D5XAN2

Thermincola

MEPYQTSIPVHYYEINNHRQASPVAILNYLEEAAIRHSESVGWGIEKLLANSRGW
71




potens

LLTRWSLHMQKYPQWGETVNIETWPYKFERFYATREFRISDREGRVLGAATTLW




(strain
VFFDLRRKRPVRIPPDIYDAYGTGAERMVADEFADLPVVDEPEIKLEFRVRLSDID




JR)
TNDHVNNTKYVEWLLETVPLSVHNGFLLSSVEIAYKKETSYGSTVLCGIKETEAG





ERQTTFLHIILDKDSGTELARARTVWQKRSKI






Q8XH69

Clostridium

MSENKFMKKYDVLYYDSDVNENIRMVPLMKIFGDVSAIHEEELAYEGIKYLKDH
72




perfringens

ELSWIIYSYSIDIKKPIPYKSSINVETYLEGIKKFYACRVYKVYNEKNELVAEGKIIF




(strain
LLIDLEKRRAVRIPKEYCELINMSDTGEVELKSTKVEKLIREDLESNISVRRSDIDF




13/Type
NKHVNNTKYLEWTMEATPECILDEYSLISAKIKYEKEVRLGDDVNIICQWDEIEE




A)
GYKCLYKIVNNRLGEVSASIETIWKKEF






B5B3P5

Solanum

MAEFHEVELKVRDYELDQYGVVNNAIYASYCQHGRHELLERIGISADEVARSGD
73




lycopersicum

ALALTELSLKYLAPLRSGDRFVVKARISDSSAARLFFEHFIFKLPDQEPILEARGIA




(Tomato)
VWLNKSYRPVRIPAEFRSKFVQFLRQEASN




(Lycopersicon






esculentum)








D4KXX4

Roseburia

MYSFSNRVRYSEVNSEKELTLPSLLDYLQDCCTFESEDFGVGVDYLAKEQVAWIL
74




intestinalis

SSWEIKVYRYPQMGQHIKVSTWPYAFRGFYGYRNFCIEGEDGEIFAEANSVWVF




XB6B4
MDTEKMRPARVSERMQEVYIPEIRDEIPGEWADRKISLPDEAVQKSVEKEPVRVS





RFYIDTNHHMNNGKYILVAEEYLPEQVFVCGLRAEYRKAAMLGDMLYPVVTIEE





KQITVTLADEKGASYAIICFQIQKKERQS






R7L7B6

Coralio-

MLDNIHRTSYKVRISDADQNGTLKCNALLQMLQEAATEHATILGVDFKALKPLN
75




margarita

LGWAVSKFVIDVKRLPQWGERINVTTWASDKERVATYREFVVTDSSGTELVSAR




sp.
SQWVLFDTRERKIAKMEKIQDWSRLENKYANASNFEPLKQPKTTTSSAICAARND




CAG: 312
DIDLNMHVNNAVYLIWAAESLPQNFTSVPKQIRINFLEEVMPHTNVEVLCHIDGK





NSYHTLINMNTNRECARINIFWQ






H1X5Q2

Weissella

MAEVYSMQHEVLYYEADVTGKLSLPMIFNLAVLSSTQQSVDLGVGPDYAHANG
76




confusa

VGWIILQHVVDIKRRPKIGEKVALETLAKEFNPFFAKRLYRIVDEAGNELVSIDAL




LBAE
YAMIDMEKRKMARIPQEMVDAYAPERVKKIPRQPEPDHMIGDIPVDVDQQYAVR




C39-2
YLDIDSNRHVNNSKYFDWMQDVLGPAFLEAHEPTHLNIKYEHEILLGDTVRSEAQ





IMEDKTIHRIWSGDTLSAEAHIDWTKSEN






J6HI88

Pepto-

MLKTYSENYIVDFRDVDRYYDIKIPQLLEIMGTVSTKEITNELGFDPFYLIRQGLA
77




anaerobacter

WILYEWKVDIEKTKLYAQTIKIETFAVDRKGMYFIRYFGIYDKDDKLIGRAAAKW





stomatis

VVINTTQRKIVKLPQEILEAAKINIEELNENQKYIYDMPEKPLRLEKRQDDFIQTIFP





IRFYDIDSNHHANNVKYVDWAIESLHQHEDFLKKYKVSHLSITYKKETGEDGDIIC





KTYIDNLRTYHEIYSSDNSLLTVLEFKWAERPQ






U2MZJ9

Clostridium

MGRVFEKQYEINYYNLDNSLKCKITSILDFFCDIGMQQSEELGVGVDSLTGRNLT
78




intestinale

WVFYKYNINMKRYPRYGEKIRIITDPIGFKKFYAFRDYTILDEGNKVIGEGKSLFFL




URNW
IDLAKRRPTRIPRDLYEAYGCADEIKDNPTIVDPEKLEEEQYFREFYVRRSDIDSNK





HVNNTKYVEWALEVIPDEIVDEYELDNISIVYSKEITYGHMVKTKCSVHNEDNGII





KIRHLIQDDVGRDITLINTFWRKN






F6B7F0

Desulfoto-

MLNRKYRKEFEVHYYEINQFEEATPVAVLNYLEETAVAHSESVGVGISKLKSQG
79




maculum

VAWMLNRWHIKMEKYPLWNEKIVIETWPSRFERFYATREFNIRDSYDHIIGRASS





nigrificans

LWVFLNIEKKRPLRIPDKIKDAYGTDPHRAIDEPFGELYNLDDSVEKKEFRVRRSD




(strain
IDTNNHVNNAKYVDWVLETIPAEIYHNYTLASLEVLYRKEVAFGATIWAGCQGI




14880/
DSMGKGLNPVYAHSIMNQDGNLALARTMWQRRNKNLHTN




VKM B-





2319/





CO-1-





SRS)





(Desulfoto-






maculum







carboxy-







divorans)








A7H762

Anaero-

MEKHRQTFGVHTYEVDAFGTVAIAALSGYLQEVAGQHAAALGVGLEVLMPRGL
80




myxobacter

TWVLARQRIENPVPIVLGDRLEIETWPAGIDRLAALRDFVVRRADGTEVARATTQ




sp. (strain
WFVLDLESRRPVKPAEVLDPRFQRELLPPILPLAPGKLPELRSWEFQKRFHVRYGD




Fw 109-5)
IDVNMHVTNTSYPTWAQEVVPREVFRGQRLASVEVHFLAEAHYGSAILSRLAST





GEGAFAHAIVREEDEKELARLATRWVPRAAPAVPGAR






F8F2E5

Treponema

MKALWTEQFTVRTWDVDRNNRLSPSSLFNYFQEVAGNHATELGVGKDALLRGN
81




caldarium

QAWILSRMTTLLYRRPGWGETITVRTWPRGTEKLFAIRDYDIIDGFGSTIAQGRSA




(strain
WLLVDVEKLRPLRPQSLTENLPTNTDMPAIPDGAQALTALPELQAAGTRTAAYSD




ATCC
IDYNGHVNNARYIEWIQDILDASILEQTNHFRIDINYLAEIRPQETISLWKEPLPNQ




51460/
DAGTEEHAGERPPFTPFEVTELWAFEGKHIDSGQSSFRAELRCGA




DSM





7334/





H1)





(Spirochaeta






caldaria)








C8PCP7

Lactobacillus

MEKLEKKYTIKSHDCDESGRLKMSMLISYMMDTMSHLLDPCIKIEHAGWVVVN
82




iners

YQFDINKLPKFDDQITIKIDLCYYNRFFAYIKFLVKDLQENELVTINSQWILFDLLS




DSM
RRMIELDSAKVGISDAQKIAKLPHFDRIKVLAGQEDFQRSYRVMYSDLDVNHHLT




13335
NGRYFDWIVNTIPRDFLNSHSMVAASIQFRKEILYDQSAVVTLTWNADHSVSYHT





IKRDEQILTVAKISWVSDK






Q6PUQ2

Petunia

MNEFYEVELKVRDYELDQYGVVNNAIYASYCQHCRHELLEKIGVNADAVARNG
83




integrifolia

EALALTEMTLKYLAPLRSGDRFIVKVRISDSSAARLFFEHFIFKLPDQEPILEARGT




subsp.
AVWLNKSYRPVRIPSEFRSKFVQFLRQEA




inflata







R6Q7V8

Clostridium

MLFTKEYEIKYYEQNVNGDLKESALLNFLQDIATLSAESLGFGPSFVFANNYAWV
84



sp.
VLKYHIELFAPLRNLSSIVIKTEPRGIAKLYAYRDFELYTKDGQCIGKAVSTWVLID




CAG: 306
ICTRKLLNTQKILADFMAPYQKRETDLVYEKIDSPDELMYQEVFDVRFDDIDVNR





HANNSNYIVWALETLPVDFRLKHSPKTIDIKYKKEIGINSRVLSEAQQILTDGNVQ





TLHVIKDEQNAQDLTSLKIVWQ






A0A099Y

Lactobacillus

MALKAPLVYETKEHLSYYECDATGHPSLSMLISMAVQVSEEHGNSLGLDTATIQS
85


BD8

mucosae

YGGGWVITSYEGSFAARQPINGEEVILGTRAIANNRFFALREFWIQSADRQVEYV





RLTGLFVYMNLQTRHLMSIPQAVIEPYHGPEKKRLPRLAKARELQDVRCKNDYH





VRFFDIDSNRHVNNARYFDWMQDPLGAEFLTRHQLKHMTMRYEKEVRYGQTIT





SEISSPYRHENGELTTDHRIVVDGQLAAASTMIWYE






W0ED98

Desulfito-

MKKFQKEFEVHYYEVDFYQELTPLALLNFLEETAIAHSEAVGYGVTRLKEKGYG
86




bacterium

WVLSQWQIEMDQYPHYGEKVKIQTWPSHFQRFYGDREFLVLNSQDKVIARASSL





metalli-

WIFLNLEKRRPTRIPQEVSDAYHIFPDKALSFPFPELKMSQTREKKRSKFMIRRSDI





reducens

DTNDHVNNAKYIEWVLETIPEEVYRTYRISSLEVVYKKESTYGQMIQSVTEEQQR




DSM
VDQEAHYVHQILERDGEEEVALALAQTRWSKR




15288







C7XWA4

Lactobacillus

MAGQKFILEHQVNYYECDPSGHLSLSMLVALMILASEKQNAQLGVDEHVTKELG
87




coleohominis

GGWVIIDYEGHFQREWPKENEQIKFETAIVAYNKYFVVRQFVIRDYHEQIIGTVN




101-
GLFVYMDLSKRRMAKIPEAIMTPYEAASTLRLPKVARPDKVEATDEWRENHYQV




4-CHN
RYFDIDYNGHVNNARYFDWMLDTLDHDFLLKHQIMEIRMNYEHEVRPQTEVNS





MAITIENNAHEWTTQHQIWVGTQKCATATIKWR






J9W2E3

Lactobacillus

MSMPAHSFSELHTIPFYECNVNNRISIPMLINILILASEHQNENLGLDQTYLIDHYG
88




buchneri

VGWVVTSYSIHITHLPRKDSVVKMTTRGTSYNRYFAFREFWLHDQAGNELVKVE




CD034
SIWVLMNEQTRKITPIDETIIAPYQSEKVKRVPRLARPERIEATDDVSAKEYQVRW





SDIDFNGHVNNSRYPEWMLDSLPMDFLNQHEPTNIDIRFENEVKYGNRVTSSVLV





DSSDNAKIKTVHEIKSNDVLSASATIVWKDIKAKGND






Q8L6B 1

Triticum

MGSLLEDGLSYKESFIVRCYEVGINKTATVETIANLLQEVGCNHAQSVGFSTDGF
89




aestivum

ATTTTMRELGLIWVTNRMHIEIYKYPAWGDVVEIETWCQADGKIGTRRDWILKD




(Wheat)
LANGEVIGRATSKWVMMNQNTRRLQRVSDEVRDEVFIHCPKSPRLAFPEENNGS





LKKIPVLTDPAQHSRLGLVPRRADLDMNQHVNNVTYIGWVLESIPQDIIDTHELQ





TITLDYRRECQHDDIVDSLTYIEEGEEINSNGSLFSAPHPEEQRQFLHCLRFAGAGD





EINRGRTVWRKLAR






F2JL78

Cellulo-

MDNTFSKKYTIEIYDVNSNYRCKYSSLMNYLWDVVVSQSDSLGETDNGLINNCA
90




silyticum

WVLLKYDLTIIEYPKFRDTITVETDIVGIKKLYGYRSFTIKTSEGTLIASGISTAVLID





lentocellum

INKRRPVRISPEQCKLYGIEKELEENIPLDDFIQLEGYKYSKDYRARHSDIDINQHV




(strain
NNVKYLEMAVDTLPRTILNASEISNIKVLYKKEALDEASLHVCSDVIENEKGHLTT




ATCC
LHTIIDLTHDKLLTKLELKWRKI




49066/





DSM





5427/





NCIMB





11756/





RHM5)





(Clostridium






lentocellum)








E6LHJ1

Enterococcus

MAKEFSRQHEVVYYECDMNGNMTLPTVISLAIQVSETQSNELNRGSEYIHQHGV
91




italicus

TWILTNYHLEITRLPKVDEQIIVTTKAEEYNKYFCYRSFWIRTLSGEELVHIQAVFG




DSM
LMNIETRKLSRVIDEIIAPFESQKITKIKRFGKLEKIVIGESLPYRVRFFDIDSNLHVN




15952
NAVYFHWILDVLGRDFLTSYVPKTITIRYDKEVEYGNEITSVVEKINQDQVMCTR





HAIMLHEETCCEALIEWKELSK






C5R921

Weissella

MAVEFRMPHDVVYYEADVTGKLSLPMIYNLAILSSTQQAIDLNIGPEYTHAKGLG
92




parames-

WVVLQQLVTINRRPKDGETITLATKAKQFNPFFAKREYRLIDAAGNDLVIMDGLF





enteroides

SMIDMNKRKLARIPKDMAEAYQPEHVRKIPRAPEVTPFDETREADFVQDYFVRYL




ATCC
DIDSNHHVNNSKYAEWMSDVLPVEFLTSHEPTAMNIKYEHEVLYGNKIKSEVQL




33313
VDNVTKHRIWFGDVLSAEATIEWTTASN






Q8XH68

Clostridium

MGKAYEKVYEVTYGETDGRKDCRITSMMNFFSDCCLSQEEKNSMNYADNSSET
93




perfringens

TWVFFDYEIIVNRYPRYREKIKVKTYVESIRKFYSNRVFEAYDMDGALVARADVL




(strain
AFLINKKTRRPARISDEEYEIHGLSKESSKLLRKKLNFEKFDKEDLDMKFHIRYLDI




13/Type
DLNMHVSNIKYVEWILETVPVDIVLNYKMKKIKIKFEKEITYGHNVIIKSKIIKGED




A)
EVKVLHKVENEEGESITLAETYWY






A0A0D2S

Gossypium

MLKFSYCNATDLNQALVQCRFAGSFGPLSSRRRSPRAAVSCSRSNLTPIQAVLSC
94


4B4

raimondii

QQQVGSDPVESELGSLADRLRLGGLTEDGLSYKEKFIIRCYEVGINKTATIETIANL




(New
LQEVGGNHAQSVGFSRDGFATSPTMRKLHLIWVTARMHIEVYKYPAWSDVIEIET




World
WCQNEGRIGTRRDWILKDVATGEVIGRATSKWVMMNEWIRRLQKVSDDVKEEY




cotton)
LVFCPREPRLAFPEENNKSLKKISKLEDPVQYSRLGLMPRRADLDMNQHVNNVT





YIGWVLESMPEEIIDTHELQTITLDYRRECQRDDVVDSLTGPELVEGSKIHGTNGS





ATAITREDDLDCHQFLHLLRLSSDGQEINRGRTEWRKKPT






A9RDN5

Physcomi

VLAAIASVALAAETQRRHEVFSGKTRVPVDALRQGRLVESRLVYRQTFVIRSYEI
95




trella

GADRTASIETMMNHFQETALNHVWMSGIAGDGFGATRAMSCRNLIWVVSRMQV





patens

HVEQYPAWGDAVEMDTWVAASGKNGMRRDWLVRDYKTGQILARATSTWVM




subsp.
MHRKTRKLSKMPEEVRTEISPYFLDRSAIKHESMLTQKIIRLDGNAEFVRSGLTPR





patens

RSDLDMNQHVNNVKYIGWMMESVPPTILDNYELVSMNLEYRRECGQSDVVQSM




(Moss)
ASLEPSTSGSLDVGFLQFVHLLRMESDGAEIVRGRTCWRPK






B1MVT0

Leuconostoc

MNSFEIKRRVEYYEADVTGKLALPMILNWAVLASKLQSDALGVGQSTHLARGLG
96




citreum

WIILQYEVHITRRPAVNEEITIQTYAAKYNPFFVRRPFAFFDAQGEEIIRVDSIWTMI




(strain
DINNRRMARLPQDIVDKYQAERVKQIPRMPNPIKILPNDDMISKDYHVRYLDIDA




KM20)
NQHVNNSKYFEWMQDVVPTEFLETHEITSINLKYENEIHLGHTIQSQVVLGNQSS





KHRIMLGDVVSAEAEFNWRNVTL






I0K660

Fibrella

MAAFPNALRRKFAGMAFIQTDAYTLRNYECDAAGRLSIPALMNLMQESANRNA
97




aestuarina

YDYGIDSETLQANGLGWMLMRFGLVMHHYPRSGQTIRIVTYPTGVEKFFVYRDF




BUZ 2
RVYADAVLLAEATSTWLVFDSHKRTMVPTPDFIRSLVCPDVDQPSPRLPLKPNYP





SVEVAEEAQAVTVGWFDIDSNQHVNNVVYIRWLLEQLPDAVLQTQELAELDVV





YRNETHWHERVLVQHQADDAGTFHHRLALAETGKDVLLARTRWRR






G9Z4R4

Yokenella

MNFKYVFRWHLPFLFLILLTFRTAAADTLLVLGDSLSAGYRMAAASAWPALLND
98




regensburgei

KWQPKMTVINASISGDTSQQGLARLPALLKQHQPRWVLVELGGNDGLRGFPPQQ




ATCC
TEQTLRTVLQQLEAAKVQPLLMQIRLPANYGRRYNEAFSAIYPKLAGEFNIPLLPF




43003
FMEEVYLKPQWMQDDGIHPNRDAQPFIADWMATRLAPLVKHDS






C5VPS2

Clostridium

MSGVITEKEYEIHYYETHTKHQATITNIIDFFTDVATFQSEKLGVGIDFMMENKMA
99




botulinum

WMLYKWDINVHRYPKYREKIIVVTEPYAIKKFYAYRKFYILDENRNVIATAKSV




D str.
WLLIHIEKRKPLKISSEIIKAYNLTDKKSDIKIEKLGKLPEEYTSLEFRVRYSDIDTN




1873
GHVNNEKYAAWMLESLPRNIISEYTLINIKITYKKETLYGENIRVLTGIKESEDKLV





FIHNVIRENGELLTEGETVWKK






A1ZMT4

Alicroscilla

MCDLIIEDAKNIYLWYMEKNQTTQLPKIWLDFEVRAYEVDIYNRVSPVTIANYLQ
100




marina

EAAGQHADHLGVGVTDLLKHRLTWVLTRIKIDMQQYPSRYEPVRVLTYPIGYDK




ATCC
YFVYRNFQLYNAQGKQIGQATSTWAVMDIQARKMVGVPQLITSLPIPDDEDFITR




23134
TKGKIAKVNAPLSETLFRVRWNDLDTNQHTNNAYYLQWAIESLPEEVLKSRQLA





SIDLLYRLETTWKEGVVARTEQTSTQPLSFIHQLIRESDQKELAQATTVWV






A0A0C1Y

Noviherb

MPNDLIRFGRSVRSLQIITALMLALMTNWAYSASKTVLVLGDSLAAEYGLARGS
101


HD1

aspirillum

GWAALLEKRLNAEKLDTRIINASISGETTSGGKARLPALLEQHRPAIVIIELGANDG





autotrophicum

LRGLPVASAKANLRTMIAAVRKAHAQPLLVGMQIPPNYGRQYTERFSSMYKELS





GELDVPLVPFLLDGVADNPQLFQADRLHPLAEAQPIILDNIWPHLKPLLRKQARH






E2XNS9
Pseudom
MRMWFLSAGLALLCVAQSAAAGTILIVGDSISAGFGLDTRKGWVALLEQRLKKE
102



onas
GFDDKVVNASISGDTSAGGLARLPAALAEHKPDVVVIELGGNDGLRGQPPAQLQ




fluoresce
QNLASMIDQSRAGGAKVLLLGMQLPPNYGPRYTKAFAEVFGTLAKEKDIPLVPFF




ns WH6
LEGIGGHPELMQADQLHPAVAAQGKLLENVWPALKPLL






A0A081B

Lactobacillus

MERNDLAALLYTENHEVPYYECDVTNRMTPAMILNTIILISEHQNIELGLGIDFLD
103


K12

oryzae

KFNLGWVVVQYEIDIERMPVMNETIAISTQATSYNRFFAFREFWIKDSNGETLVH




KM
VKSTWVTMDRTARKMVSIPEAVILPYQSEAVKRMPRLKRPTNINESDDLIKKPYQ




18671
VRYYDIDGNGHVNNAHYLEWLTDVLPMDFLTTHEPKQISLRFENEVQYGHMIES





QVTKPVESEGSMVTHHQIVVEDTISATATIEWRSRVE






C6Q1L2
Clostridium
MKKVETEKQYEIQYYEIDCNKKLLLTSLMNYLEDACTMQSEDIGIGLDYMKSKK
104




carboxidi-

VAWVLYKWNIHIYRYPLYREKVKVKTIPESFRKFYAYRSFQVFDSRGNIIADASSI





vorans P7

WFLINTERRKAMTVTEDMYEAFGLSKEDNKPLSVKKIRKQERVDSEKVFSVRYS





DIDTNRHVNNVKYVDWAVETVPLDIVTNCKIVDIIIAYEKETTYGAMIKVLTQIDK





KEEGFVCLHKIVDEEDKELALIETLWKNEK






G6CEI8

Lactobacillus

MPGKQYSENYQIPYFETDIKGELTLASLVNILILASEHQLNALNVGEATMHALNL
105




curvatus

GWVVTQYQMKITRMPKVDEKVRIVTEAESYNKYFCYRNFWLYDEAGNECVFVQ




CRL 705
SIFVMMSYETRSMVQVVPEIMVPFESSEIKGSKRFPRIKKIDPKQVTTKEYRVRYF





DIDGNQHVNNVHYFEWMLDALDYDFLTTHRVASVNIRYGHEIQYGQMTQSMVE





QLIVDDIITTRHKVAVDDLSAAEAEITWKER






R5RBT8

Proteobacteria

MELKKYTKEYTIRSYECDRNNNLRIVTLMNIFQDMADINAAQLGLGLDYVLSKG
106




bacterium

FAWVGSNYEIRIKRLPKIHEKVKIVTWPAVEKKLAAIRDYEVYGKDGERIIAASSQ




CAG: 495
WILINFMKKRPISLRDNLPEYQIIDDRAIETEFEGKIKEVERIDEQTKFRVRFDDIDL





NKHVNNGVYALWASEAVNPDFRLSHNPSKIEINYKKEGHIGEKITVLTECDGLVT





THSIQTYDGDNRELARARIEWAENEE






A0A0871

Vibrio

MVRLFSLLIMFFLSNVAHATEKVLILGDSLSAGYNMSAEQAWPNLLPEALNTYG
107


QH8

vulnificus

KNVEVINASISGDTTGNGLSRLPELLKTHSPDWVLIELGANDGLRGFPHKVISSNL





SRMIQLSKASDAKVALMQIRVPPNYGKRYTDAFVELYPTLAEHHQVPLFPFFLEE





VIVKPEWMMPDGLHPMPEAQPWIAQFVAKTFYKHL






R6XTQ7

Alistipes

MEKINVSGNRFSTDITIPCYDTDASFRLKPAAFMDHAQEMAYLAAQALHFGYDD
108



sp.
LQRHHTAWVLSRMRMDFLNPPKWTDETTLYTWHKGQDGLFFLRDFELRRKGDT




CAG: 435
DFADKSKAQVLCTSSWIVMNVETRRLVRSDEVLNMVPATTQCPDNAIQIPCGKV





VMPKNIPAEEVGCHKAAYSDIDVLGHTNNARYVVWAMDCIDYEEVAGNPIRSISI





NFIKETKPGEVVRIFRSVEDIDGQKKYFIEGKIEDKPCFCARIDF






A0A011R

Alkali-

MSLVYKSEKTITHYMCDRSRSLTLPMLVNLLLEVSEEQSSELSRDESYLKARGVN
109


NY4

bacterium sp.

WIILRYEFSVSRMPNLKETINIETRASEYNKLFTYREFVVKDSSGKVLLTVDTTFAL




AK22
MDLSTRKMVRLTDEIVSPYQATASRRIRRSDKPKELTDFDDCKQRTFDVRYFDID





GNGHVNNAHYISWLLDSLPSDFLKSHEVSWGVIAFDKEVSEHQSIDSLSMRRKER





GTATDHQIKSEAAVHCKASFTWKKLNKEEH






Q4R7D6

Macaca

MARPGLIHSSPGLPDTCALLQPPAASASAAPSMSGPDVETPSAIQICRIMRPDDAN
110




fascicularis

VAGNVHGGTILKMIEEAGAIISTRHCNSQNGERCVAALARVERTDFLSPMCIGEV




(Crab-eating
AHVSAEITYTSKHSVEVQVNVMSENILTGAKKLTNKATLWYVPLSLKNVDKVLE




macaque)
VPPVVYSRQEQEEEGRKRYEAQKLERMETKWRNGDIVQPVLNPEPNTVSYSQSS




(Cynomol
LIHLVGPSDCTLHGFVHGGVTMKLMDEVAGIVAARHCKTNIVTASVDAINFHDKI




gus
RKGCVITISGRMTFTSNKSMEIEVLVDADPVVDSSQKRYRAASAFFTYVSLSQEG




monkey)
RSLPVPQLVPETEDEKKRFEEGKGRYLQMKAKRQGHTEPRVAMATGPVSTQKFP





PWPKTRFTLRAGIV






A9PJK8

Populus

MVATAAASSFFPVPSPSGDAKASKFGSVSASLGGIKTKSASSGALQVNTNGQAPP
111




trichocar

KINGPPVGLAASVETLKNEDVVSSPAPRTFINQLPDWSMLLAAITTMFLAAEKQW





pax

MMLDWKPKRPDMLIDPFGIGRIVQDGLVFRQNFSIRSYEIGADRTASIETLMNHLQ





Populus

ETALNHVKTAGLLGDGFGATPEMSKRNLIWVVTRMQILVDRYPTWGDVVQVDT





deltoides

WVSASGKNGMRRDWLVRDAKTGETLTRASSVWVMMNKVTRRLAKIPEEVRGEI





EPYFLTSDPVVIEDSRKLPKIDDNTADYICESLTPRWNDLDVNQHVNNVKYIGWIL





ESAPPPIMESHELAAITLEYRRECGRDSVLQSLTAVSDTGIGNLGNPGEVEFQHLL





RFEEGAEIVRGRTEWRPKHADNFGIMGHIPAESA






A8MEW2

Alkaliphilus

MARYTEEFVIPYYDCSGDRFVRPESLLEYMGEASLLHGDTLGVGGADLFKMGFA
112




oremlandii

WMLNRWKVRFIEYPKSRTTITVETWSSGVDRFYATREFNIYDSDRKLLVQASTQ




(strain
WVFCHILKRKPARVPDIISAVYDSEDEHNFYHFHDFKDEVQADEAIEFRVRKSDID




OhILAs)
FNHHVNNVKYLNWMLEVLPKQFEDQYLYELDIQYKKEIKQGSLIKSEVSMDIEG




(Clostridium
EETVCYHKITSNSVLHAFGRSVWKNRK





oremlandii






(strain





OhILAs))







O00154

Homo

MKLLARALRLCEFGRQASSRRLVAGQGCVGPRRGCCAPVQVVGPRADLPPCGA
113




sapiens

CITGRIMRPDDANVAGNVHGGTILKMIEEAGAIISTRHCNSQNGERCVAALARVE




(Human)
RTDFLSPMCIGEVAHVSAEITYTSKHSVEVQVNVMSENILTGAKKLTNKATLWY





VPLSLKNVDKVLEVPPVVYSRQEQEEEGRKRYEAQKLERMETKWRNGDIVQPVL





NPEPNTVSYSQSSLIHLVGPSDCTLHGFVHGGVTMKLMDEVAGIVAARHCKTNIV





TASVDAINFHDKIRKGCVITISGRMTFTSNKSMEIEVLVDADPVVDSSQKRYRAAS





AFFTYVSLSQEGRSLPVPQLVPETEDEKKRFEEGKGRYLQMKAKRQGHAEPQP






Q14814

Bos

MSGPAAETPSAIQICRIMRPDDANVAGNVHGGTVLKMIEEAGAIISTRHCNSQNG
114




taurus

ERCVAALARVERTDFLSPMCIGEVAHVSAEITYTSKHSVEVQVHVMSENILTGTK




(Bovine)
KLTNKATLWYVPLSLKNVDKVLEVPPVVYSRQEQEEEGRKRYEAQKLERMETK





WRNGDIIQPVLNPEPNTVSYSQSSLIHLVGPSDCTLHGFVHGGVTMKLMDEVAGI





VAARHCKTNIVTASVDAINFHDKIRKGCVITISGRMTFTSNKSMEIEVLVDADPVV





NNFVKRYRAASAFFTYVSLSPEGKSLPVPQLVPETEDEKKRFEEGKGRYLQMKA





KRQGQAETQA






P34419

Caenorhab-

MVGHSESSTDAVIEPTSEELLAEQVRVFNKMKGSTNFNRVAEDVYPVEVTKSKL
115




ditis

VCEMVVQHQHLNSKGTLHGGQTATLTDVITARAVGVTVKDKGMASVELAVSYL





elegans

LPVKVGDVLEITAHVLKVGRTMAFTDCEFRRKSDGKMSAKGKEITLAFLPNQPGI





SVENGTQF






R5BT85

Blautia

MGYQFRSRVRYSEIDEDGKLTLPAILNYFQDCCTFHSEDVGLGMKKLRKIHRGW
116




hydrogen

VLSSWQIIVERYPEHGEELTVETWPYDFKGFMGMRNFILRTSQGESLCKANTLWS





otrophica

FMNTDSGMPVKLQPENTQGYQLEPKLEMEYAPRKIGLLSQGEKRESFLVQKHHL




CAG: 147
DTNHHVNNSQYITMATEYLPKDFEIWQMRAEYKMQARLGERIIPWVSEEPKRCV





VSLNQETGKPYAIVEFSKKEK






DOBKNO

Granulic

MTVDVICQIERTILPYECDWKENLLLSQALGMMMLASRKQQQQLQNPNLIYEKG
117




atella

YTWIVIQHEIEIQRMPKVDEEVIIETQAISYNKFFTYREYRILSKEREELFKCITTFA





elegans

MLDMKARKIVSIDEEVVLEYPLSIGKEMRKATRIPKKDFSDATTGEYKIRINDIDA




ATCC
NLHVNNARYFDFAFSELGMEFIEDHQLKQVVIKYEKEVLPESTISCSTLWEENTLE




700633
SQERRQTYHLISQDGNRCANIQMKWEEIV






R5EEV3

Firmicutes

MEPIFQQDFPVQELCVDRYGRLKPSTLLYFAQEIAGRHCDELADTLESHRLFWAV
118




bacterium

TRHRVQINRLPELGETVHIETWPMPNTHVGYPRSIVIYDQAGNECSRSISLWVLM




CAG: 110
DQDTRSSVSPDKSGIIVPGTLRGTELALPGGLVPRAMEHSCQRDVCFTDLDRNGH





MNNTRYMDWIDDLLPSDFHREHPVKEFAVRYHSEAREGQRLDLHWDFVEDNCL





RVDARRRNETRDELVFSAKVLFD






R5NTL8

Clostridium

MKLENSIFEETYRTSFSQTGIHETLTNKSFLSMMENLAGAHSGYCHYSFANLAPE
119



sp.
HKTWIILNWKLQVFRRPYADEIVTLKTWGHFANKIYVLRDFKMLDKDGNLLAIA




CAG: 793
SSKWCLFDFSTGRIARLPDNLEEIYQGFNSESVFNCNDLPKLKAPESEPIASDTYKI





RRFDLDLNKHVHNLNYLNIAYELLPLDVYDGPELNNVEIVYKKEIKYGDTIKSYL





YKENDSYIIVIKSLDGSIVHSIVKLY






Q1J6W5

Streptococcus

MGLSYQEELTLPFELCDVKSDIKLPLLLDYCLMVSGRQSAQLGRSNNNLLVDYKL
120




pyogenes

VWIVTDYEITIHRLPHFQETITIETKALSYNKFFCYRQFYIYDQEGCLLVDILAYFA




serotype
LLNPDTRKVATIPEDLVAPFKTDFVKKLHRAPKMPLLEQSIDRDYYVRYFDIDMN




M4
GHVNNSKYLDWMYDVLGCEFLKTHQPLKMTLKYVKEVSPGGQITSSYHLDQLT




(strain
SYHQITSDGQLNAQAMIEWRAIKQTESETD




MGAS10





750)







R6FXC3

Clostridium

MGISYEKMYEIHYYECDKNLNCTLESIMNFLGDVGNKHAESLNVGMEYLTERNL
121



sp.
TWVFYKYNIKINRYPKYEEKIKVKTVAEEFKKFYALRTYEIYDENNIKIVEGSALF




CAG: 221
LLIDIVKRRAVKITDDQYKAYNVDKGSTGKNLIGRLERLEKVKNNEYVSNFKVR





YSDIDFNKHVNNVKYVQWFMDSVPQEIREEYELKEIDILFEHECYYNDEIKCVCEI





HKNEDNLLVLSNIQDKDGKELTVFVSKWE






R7F611

Clostridium

MGYFEHDFEIGLRDVENPNYLSNKAILAFFENIGSYHSDSINFGLNEIPKTKSSWVL
122



sp.
LGWKVKVLKRPLYGDKLHIVTWARNTEKFSTYRDYEVYNQNNELVIIGTSKWVL




CAG:354
VNTTTGKLRPIPEEIIKLYCPDTKTAFPPEEALLTKLTDSEHYGTACTCTVGRSQID





LNNHMHNLYYLDMAYEALPEEVYKNNTFNFFEITYKKQIRLHDAVKCYYVFEEN





THKVVIKSLDDKKTHAIIVFK






C4JAL9

Zea mays

MHHRFAGLVPTARPALPPIHGGVVGRSYPPVHRSLALRLAPFASASVRRACRPLA
123



(Maize)
VSAQSTSLRPEKFFEVEMKVRDYEIDQYGVVNNAIYASYCQHGRHELLESVGISA





DAVARSGESLALSELNLKYFAPLRSGDKFVVKVRLAGIKGVRMIFDHIITKLPNHE





LILEAKATAVCLNKDYYPTRIPRELLSKMQLFLPVDSRGSNEDVNNRNNSCN






N1JPQ7

Mesotoga

MKPVVTKEVYRVRYYELDCQWKASISSLMDYFNDVVTLQTVEMGHGVDVMSK
124




infera

GEYAWLLLRWDVKVNRYPDYLENVVVQTIPYSMDRFYAYRRFEIFDCSGNVIVD





ANSQWILIDQRKRRPIRIGDQFYELYGIDSDFHEPLSFPKVNENESSSEEITFIVRNS





DLDTNGHSNNVAYVRWIMETVPSEFVKRFLKRLTIEYKRESRKGDVISIESVFENG





AEFAEGKHKITSSGRVLSLARTEWK






B1ZXQ1

Opitutus

MPEKLTLNASVLYADVDRTEVLLLRGVFKFLQEAAITHANQFDLGSRAMATRGE
125




terrae

SWVLNRMAVAVHRYPRYEETMRIETWSRGIKGFKGYREFRVFDAQGAPLFSGSS




(strain
LWLYVNMRTKSIIRVPAELAAEFPKRDDGAFFPELESLEFAPPAADARRVPIAIRY




DSM
SDVDVNAHVNNTAYLDFLQEALARAGLSPRPQSIRIKYARAIPAEAETVRVAIEPR




11246/
GTGAAFAIEDHDTIFAIGEVD




JCM





15787/





PB90-1)







E4MXK9

Eutrema

MVMTHCTRFQHLLQPKLLFSHSRVFRHPHIRARTPLRSIMGSSSSFSSKLLFRQLFE
126




halophilum

KESSTYTYLLADVSHPDKPALLIDPVDKTVDRDLKLVNELGLKLIYAMNTHVHA




(Salt
DHVTGTGLLKKKVPGVKSVISKASGSKADMFLEPGDKVTIGDLYLEVRATPGHT




cress)
AGCVTYVTGEEADQPQPRMAFTGDAVLIRGCGRTDFQGGSSDQLYESVHSQIFTL




(Sisymbrium
PKDTLIYPAHDYKGYEVSTVGEEMQHNPRLTKDKETFKTIMSNLNLAYPKMIDV





halophilum)

AVPANMVCGLQE






A0A089R

Cedecea

MMNFNNVFRWHLPFLFLALMTFRAAAADTLLILGDSLSAGYRMAATSAWPALL
127


LX5

neteri

DAKWQPQNTKVVNASISGDTAAQGLSRLPALLKQHQPRWVLVELGGNDGLRGF





QPQEVEKTLKQVITDVKAANAQPLLMQIRLPANYGRRYNEAFSAIYPQLAKQFDI





PLLPFFMEEVYLKPQWMQDDGIHPNRDAQPFIADWMATQLTPLLSK






J5XTZ 9

Klebsiella

MMNFKYVFRWHLPFLFLVLFTCRAMAADTLLVLGDSLSAGYRMAGNTAWPALL
128



sp.
NDKWQTKTPVVNASISGDTSQQGLARLPALLKQHQPRWVLVELGGNDGLRGFPP




OBRC7
QQTEQTLRTIIEHIKAANAQPLLMQIRLPANYGRRYNEAFSAIYPALAKEFDIPLLP





FFMEEVYLKPQWMQDDGIHPNRDAQPFIADWMATRLAPLVNHDS






M4VN84

Bdello-

MSTSSETTEKRIWEEEYKITSYLVNLRGRAGLYAILNLIQDVGWMHAIAAQVRLP
129




vibrio

ANLAWVFTRQKLVMSQWPKWNETISIRTWLRPPESAAFILRDYEIILNGQVIGTCT





exovorus

STFAVIDTQTRKIAAQEWSEYEQLFRTGTALPHHPVKIPYREDAQDLTVFEVRNSD




JSS
IDLNNHVNNTKYAKWILDSISIDTLRAGVDLLEYEVNFLAEARSGDRVTVQSCAE





EKLEGQSDSATALIQFQGVRVSDKKTIFTAKLRVR






L7VMU3

Clostridium

MIPELVYRNSYIVGYRDVDFNNDLRLSSLFGYFQDTAIMNVEKLGIGVNTLSEKY
130




stercorarium

SVSWVLTKILVEINRIPKWNEKITVETWPHRPKKFEFDRDFRVRDDNGNIIAAAIS




subsp.
NWVLLDLKTREIRKSEIISGDYPPLEFTDERALEGRLRKLRPAGEPEVVYKRVLGY





stercorarium

SDTDANGHINNAKYIDFIMDCFSIEEHKKHSVRSIQVNYLKEVFPGDTLILYRDVS




(strain
GAGSNQVYIEGINEADQKPAFSAELKFD




ATCC





35414/





DSM





8532/





NCIMB





11754)







A0A0C1Q

Clostridium

MKNIHRENYKVKFNETDYSTKIKMHSLINYMQETSSIHAELLGAGYEELKKHNLF
131


ZB7

argentinense

WVVSRLKINMKKYVNWNDEVIVETWPSGVDKMFFTRSFRIYDREENHIGDINAA




CDC
YLLVAEDSMFPQRISKLPINIPTIENRFEPYERLEKIKFPKDDKVLVAKKKVRYNDI




2741
DLNLHVNNAKYIEWVEDCFPLEMYKDMRIETLQLNFIKEAKCGEKIFFYKYNDLE





DENTCYIEGIEKQSESQIFQCKLTFNKL






P41903

Saccharomyces

MSASKMAMSNLEKILELVPLSPTSFVTKYLPAAPVGSKGTFGGTLVSQSLLASLH
132




cerevisiae

TVPLNFFPTSLHSYFIKGGDPRTKITYHVQNLRNGRNFIHKQVSAYQHDKLIFTSMI




(strain
LFAVQRSKEHDSLQHWETIPGLQGKQPDPHRYEEATSLFQKEVLDPQKLSRYASL




ATCC
SDRFQDATSMSKYVDAFQYGVMEYQFPKDMFYSARHTDELDYFVKVRPPITTVE




204508/
HAGDESSLHKEIHPYRIPKSITPENDARYNYVAFAYLSDSYLLLTIPYFHNLPLYCH




S288c)
SFSVSLDHTIYFHQLPHVNNWIYLKISNPRSHWDKHLVQGKYFDTQSGRIMASVS




(Baker's
QEGYVVYGSERDIRAKF




yeast)







C4Z694

Eubacterium

MVFNYTYRIGLEDCGRENKATNRAILTILEDIAGLHSATVGLGLNEINETGCAWV
133




eligens

VLNWQMKIIRRPAYNDELTVYTWSTSADKLFAERDFRITDKNGETIVIATSRWLY




(strain
MDINRRRPVRITPEIMDRYESEPEIHVFTEKVNRIDPPDTGYIEIPYNILRRDVDYLG




ATCC
HMHNISYLDAAYDVMPEEYFNGPQFNFVSIEYRKELLRNDEVKAHFYKIDKGCII




27750 /
SLNTDKINAVIMLKY




VPI C15-





48)







A0A0B3V

Terrisporo-

MIYCNNYKIGLEDIGIKNEATNKALLAIMEDVAGLHSASVGYGVLDIETKKRVWI
134


S79

bacter

LLDWKMKVIKRPKYNDDIKAETWSRKVERLYAYRDFQLKDKEGNIIAIGTSRWIL





othiniensis

IDTDRKRPMKLTADIADLYESETDKSVFPEQIEDIKCENYLFKKDYYIQRRDIDINE





HMHNLNYLDMAYEILPEDVYKNKVFDNIRIVYKKEILYGEKVVCYYEEQGNKHII





TAKSKDKINAIIELS






A0A084J

Clostridium

MELNYKEQFTIKFHEGDFMGNVKLFTIMDYVQQVSEGHSQILGVDFQSMMNKGL
135


BW2

sulfidigenes

FWVVSRVEITMERYPKVGEDITVETCLGGREKVFMKRRFKIKDKDGQVIGRVLIY





YLIVDIETRLPQKPSMCPVDININVGDVIDNKLNKIKMPGEAIETVNRKLYYNDIDI





NNHVNNAKYISFIEDFFSLDWHRVKKISYMQLNFIKEIKFDDSLIMNKFIEDKESNS





FCINGISEISEQEFFQCRLKF






A0A037Z

Clostridium

MEGLVTEKEYEIHYYEVDYKRRLLITNIINYFCDIATKQSEDRNVGLDYMKENNV
136


4L7

tetanomorphum

AWVLYKWHINVHRYPLYGEKVIVTTRPHSFRKFYAYRKFEIIDEKGKIIIEANSIW




DSM 665
FLIDIQRRRPKRINEHIEEAYKVSKDNDERAILEIPDIKCIEKIHNEKTFNVRYSDIDT





NGHVNNAKYVSWAIETVPLEIIKSYALKNITINYEKETKYGESINAFVEVIKEDKM





VICRHRITDKEGNELTIAQSTWE






A0A0L8E

Clostridium

MDKIISKKDYNIHFYEVNYDKKADITSIMSYLGDLATYQSEELGVGIDYLMRNKM
137


W05
sp.
AWVVYKWNVHMDKYPEYNDTITVTTIPYSIRKFYAYRKFEIFNKGEKIGEATSLW




DMHC
FLINTERRRPCRVPEDIYRAYGLRVEDDQQLEFEKLLLPSEISSEKSFDVRYSDIDT




10
NKHVNNVKYVSWALENIPLDVVKNCSVSSIRVIYEKETSYGETITVQTQMKEIED





KYIFDHVIKNSEGEKLTLIKTEFLKA






B8I625

Clostridium

MEPLSIYKKNYHVDYGDADFYKRLKLSYLFNYFQNIAGLHSENTNVGIRKLQND
138




celluloly-

YGAAWVMTRMLMDINRMPECNEEISIETWPVEPKKKMIDRNFIVRDMDGSILAS





ticum

AISTWVILDMEKREMVRIDSVIPPQYPEFLKSKAIDRKFDKLKPSGQLQPAYKKLV




(strain
GFSDIDINGHVNNAKYIDYILDCFTVEKHGEYRVKSIQINYVNEAVAGDIISLYKD




ATCC
TVDMDGSDKAVYITGINEVDGKVNFESHIRVQ




35319/





DSM





5812/





JCM





6584/





H10)







D5ADX2

Picea

MYNMDLFGAKGMARPFELELKVRDYELDQYGVVNNATYASYCQHCRHELCEAI
139




sitchensis

GFSPDVIARTGNALALSELSLKYLAPLRSGDSFVVTARISGSSAVRLFFEHFIYKLP




(Sitka
NREPVLEAKATAVYLDKIYRPVRLPADFKSKITLFLRNEELN




spruce)






(Pinus







sitchensis)








A0A0D3V

Paenibacillus

MGNIWTEEHLIYSNEIDYKANCRLSNLLSLMQRAADGDVEHMGGTRDQMVAHH
140


4E9
sp.
LGWMLTTIDLACERMPIFNETLKITTWNKGTKGPLWLRDFRIFDENNQEIAKACT




IHBB
LWALVDIDKRKVLRPSAYPFNINSNHEDSVGPVPDKLNISDEVELYHSYSITVRYS




10380
GIDSNGHLNNSRYADLCMDTLTQSELDTLSILGFHITYYHEVKSAEQIQVLRSDHL





EGYIYFRGQSLEDERYFEACLHVG






P0A8Z3

Escherichia

MNTTLFRWPVRVYYEDTDAGGVVYHASYVAFYERARTEMLRHHHFSQQALMA
141




coli

ERVAFVVRKMTVEYYAPARLDDMLEIQTEITSMRGTSLVFTQRIVNAENTLLNEA




(strain
EVLVVCVDPLKMKPRALPKSIVAEFKQ




K12)







A0A0M2

Catabacter

MNTKLEQLYTIRAFDVDTKGKWRPSAILTRLQEIAEDHAIAVNAGRKELVEERG
142


NEM6

hongkongensis

MAWMLTRLHLQMKQYPDLTDTIKVVTWPGKPTKLFFVRHSMFFSETGEELGRA





TSLWVLFNIRERFLCRTGDIGENYPYDLSHGRALPDPGKIKLPDEMQYMTTRTVA





YSEVDMNGHLNNAKYADWICELFDISHLKKAYMDQFRINYIAEAYMGQKVDLY





CKEIDGTWFVCGKTGNKTVFDASIQWK






R6RDZ9

Firmicutes

MNYYQKELVLQEKDFINDELSPYSILNYFQDIAGIHADKIGLSHEELIKNDLVWVV
143




bacterium

LRNKYEIIKMPSINQKVILKTWPHQKGKIDFDREYAIYDENNNLLIKGLSKWILMN




CAG: 449
YKTRRISMFNNIKYSFECLEETNFENKFNKIEDFDINNFSFIETTTSENDLDINGHVN





NASYARIVLNNIDFDITINHFEINYIKEIKANQKLKIYYLKKDNTYYIKAFNNEEVIF





VLIVY






E4L0C9

Pepto-

MKIFCKEYEVMNFLSSDGDLKLNHLVSYLIETSNYQSIDLGLSNEKLLDMGYTW
144




niphilus

MIYKWKIKINRYPRSYEKIKIKTWASGFKNINAFREFEVYCQGEKIIEASAIFLLIDV





hardi

EKRKAIKIPEVLAEIYGNNGNRIFKSIERVNEPSELEIANRFSYKILRRDLDFNNHV




ACS-146-
NNSVYLELIYEAVTDEYTHVKFKDINVNYINELKLGDEIVIDFYREEDRFYFFFKS




V-Sch2b
KDQSQIYARICGVSETPIS






A0A062X
Lactobacillus
MAENLYRMPHQIVYYETDPTGKLSLGKLVDLMMLASYAQGKDVGMPEEKLNA
145


6R3
animalis
QGYGWVITQHLLSITRLPRRDEKVVIETKATAYNRYFCYRNFYLRDEQGELLAK





MHTAFVLLDLETRKITRITSDVIAPFGPEPIRSIERSASPKRLEEVMLAKDYRVRYF





DIDSNHHVNNVHYIEWMLDVLDKDFLMEHEPVALNIKYEHELNYGQTCTSKVEL





LRSKDELTTLHEIYMADGTLSCSAQVTWK






A4A3N9

Congregi-

MGLLLGLALLLTGQLARAESTAGERPRILVVGDSISAAYGMSLEQGWAALLERR
146




bacter

LQTRWPGAQVINASISGDTSAGGARRLPKLLAEHSPDLVVIELGGNDGLRGYPTS





litoralis

KLEANLSFMAEAASTAGAEVLILPMEIPPNYGPRYTRSFRESFERAATDTGATLGP




KT71
FLLDGIATEEQLMQQDGIHPTVEAQPMITDIVQPVIEALLALREAS






B1IHP0

Clostridium

MQPVITDKNFEINYHEIDFKKRVLFTTIMNYFEDASLEQSEKLGVGLQYLKENEQ
147




botulinum

AWVLYKWNVTIDRYPEFGEKIIVRTIPLSYRKFYAYRRFQIIDKTGKVIVTGDSIWF




(strain
LIDINKRRPIKVTEDMQNAYGLSETKEEPFKIDKIKFPEEFHYNNKFKVRYSDIDTN




Okra/
LHVNNVKYISWAIETIPFDIVLNYTLKNFVITYEKEVKYGNDINVYSEMVHNDNN




Type B1)
EIVFVHKVENEEGKRVTSAKSIWVK






V5WGX9

Salinispira

MERNMKFSREYEVRGFEIDENYHLKPYHIASYFQDAMAQNFADNMLAAYDLQK
148




pacifica

EGRTWVLSDLCIDFLHQMPRWRTSVLVETWVSSIRGFRLTIDFRVSDSRGTPISQG





SSSWVIVKKPGNRPEKIEPYARKLGEPHSPLYPGYRFHEPDLDGSPSPLPYGACGL





RHPWEEDQKAWSICQPIRSYDIDFNGHVSNIRYIAGAVEAIPVELRSSLRPSSFRIK





YLREAVLDQVLVSEVRTVHEDSDAIEYHHILKECESAVEFSRMVSVWKR






A0A099R

Clostridium

MGKLVTDKEYEIHFYEVDYKGRALFTSLMNYFGDISSKQSEDRNMGIDYLKKVN
149


RK7
sp.
MAWVLYKWNVKIHRYPTYREKVIARTVPYSFRKFYAYRKFYILDIEGNVIVEADS




HMP27
LWFLIDIETRKPVRVQEEMYTGYCLSKDDNEIIDIPKITAPNESDFCKTFDVRYSDI





DTNGHVNNSKYISWILEAVPLNIVTQYSLSNLIITYEKETTYGEVIDSCVEVREVD





GKAVCKHKIVDKEGNELTVAETTWTR






R7LAU3

Coraliom

MKFSRIYNVRSSEITPEYKLKEFYIGMYFQECFAEYMASKGLAAYDLAKSGQTWL
150




argarita

TSDVQIDYLQEMPFWREPVEMQVWVRQISAIRIYVDFEAIHKSSVIARGSSIQLIAE




sp.
KSTHHPIKNATIASISSLSLIHESALPGVEFKKIDPFGGEYSKTSQVVRYDDLDFNM




CAG: 312
HLNNVKYVPRALESIPQEFRNSHTLKEYRIKFMRETFFNNTVSSEAFRDGNRIFHR





LARVEDGVELCRMESLWE






Q2BL39

Neptunii-

MRTAITFLLFVFLTLPVQARSLIVLGDSLSAAYQMAPEEGWVALLEEKMATEGYA
151




bacter

YDVINASVSGDTTQNGIARLKTLLKQVDAEIVIIELGGNDGLRGTPPFAIKRNLSRL





caesariensis

VNMAKDSGAQVLLLGIQLPSNYGAAYNKQFSEIYPVIAEDENVALVPFFMEQVA





LVPERMQDDGIHPSAEGQPYLLNTVWPHLEPLIN






Q3ADW4

Carboxy-

MNSNIFELEYRIPYYDVDYQKRTLITSLINYFNDIAFVQSENLGGIAYLTQNNLGW
152




dothermus

VLMNWDIKVDRYPRFNERVLVRTAPHSFNKFFAYRWFEIYDKNGIKIAKANSRW





hydrogen

LLINTEKRRPVKINDYLYGIYGVSYENNNILPIEEPQKLLSIDIEKQFEVRYSDLDSN





oformans

GHVNNVKYVVWALDTVPLEIISNYSLQRLKVKYEKEVTYGKTVRVLTGILSEQK




(strain
TIVSLHKIVDEDETELCFLESVWFLNEKLS




ATCC





BAA-161/





DSM





6008/Z-





2901)







Q5SJV0

Thermus

MESVTRIKVRYAETDQMGVVHHSVYAVYLEAARVDFLERAGLPYHRVEARGVF
153




thermophilus

FPVVELGLTFRAPARFGEVVEVRTRLAELSSRALLFRYRVEREGVLLAEGFTRHL




(strain
CQVGERAARIPEDIYRALSVLHLK




HB8/





ATCC





27634/





DSM





579)







E3G686

Enterobacter

MPFLFLVLLTCRAAFADTVLVLGDSLSAGYRMAANAAWPALLNAKWQPKTDVI
154




lignolyticus

NASISGDTSQQGLARLPELLKQHQPRWVLVELGGNDGLRGFPPQQTEATLRAILK




(strain
DIEAANAKPLLMQIRLPANYGRRYNASFSAIYPKLASEFNIPLLPFFMEEVYLKPQ




SCF1)
WMQDDGIHPNRDAQPFIADWMATRLAPLVNHDS






E6VY45

Pseudo-

MTAHLPLTHDRLYRIRSYEPRPDGLAPITAICNQLQDIASGHADALGFGYHDLETG
155




desulfovibrio

GHFWLLARLHVMMDRLPAYGGAVRVQTWPSGNERLVANRDFLILDPAAAQETV




aespoeensis
MGRATSSWVTMNASTHRPESPSEVLSTRFIPDRERALTFPAKSITRLKDGEHETGL




(strain
TARRADLDINGHVNNVRYAELCLEAVPQAWEAAHRCLGLDIQFRSESFAGDAYV




ATCC
SACAEAGPDSGMRTLLHRLTRINDDREIVRMRSWWQTG




700646/





DSM





10631 /





Aspo-2)





(Desulfo-






vibrio






aespoeensis)







A0A099L

Vibrio

MVRLFSLMLMLILSWTATAAEKILILGDSLSAGYNMPAEQAWPSLLPDVLKTYG
156


W85

navarrensis

KDVQVINASISGDTTGNGLARLPDLLTTHSPDWVLIELGANDGLRGFPPKTIAANL





SRMIQITKAAGAKPVLMQIRVPPNYGKRYSQAFFDLYPTLAEHQQVPLLPFFLEQ





VITKPEWMMQDGLHPTADAQPWIAEFVAEMFSQHL






A0A0D6Q

Anaero-

METFKESFAVHSYEVDAFGTLAPPALTGFLMEAAGLHAGRLGVGIDALMEKGLT
157


P57

myxobacter

WVLVRQRTEMPVPIVLGDVLEVETWPVGVDRLAALRDFVVRRRDGAEVARGTT




sp. PSR-1
QWFVLDVKIRKPVRPETALDARFPRELGKPVIDVAPGKLPELRTWEFQKRFHVR





YQDIDLNLHVNNGSYVAWALEAIPKDVYTGSRVAALEVQYLAECHYGSAVLSRL





ARTGPGAFAHAIVREEDEKELARITTSWVPR






N6Z483

Thauera

MPLRSIATFFFMLLFVGAAHAATILVWGDSLSAGYGLEPGRAWPTLLQTRLQEKG
158




linaloolentis

FRHTVVNASVSGETSAGGRSRLPAALERHKPDIVILELGANDGLRGLRPQLMAEN




47Lol =
LEAMIAASRDAGAQILLVGMQMPPNYGPAYTRRFAQTFDDVAKAQQVPLVPFLL




DSM
EGFAGQPERFQADGIHPTADAQPLVLDTVWRGLEPLLKRN




12138







A0A094JF

Shewanella

MLVILLTAPAQAATLLIVGDSLGASYGVNEKDGWVEGLRNALPQHTLINASVSGE
159


G0

mangrovi

TSGGGLRRLPSLLSSASPDVVLIELGGNDGLRGFPPQQLKNNLTKMIALAKQAGA





KVMLSEVMVPPNYGPRYEKAFTSVYQQLAEDKSVTLVPFFMTVIAPHPELMQRD





GIHPNTVAQPKITAFMLPFIKHALDEVNNS






A9SDT6

Physcomi-

GGFVDNSLMYRQIFVVRSYEVGPDRLMSIREIFSLFQETALNHVQLLGIAGDGFGA
160




trella

TRAMNRLGLIWVVIKMKVEVNRYPVWPEVVEIDTWVAHAGKNGMQRDWIMR





patens

SYQTDEVLARATSTWCMMDGVTRRLSKIPDEVRAEIVPCFMDDYPSSFREDEESP




subsp.
RITKLDNTTAENRRSHLKSTTADLDMNQHVNNLKYINWVLDSVPVEHMEKHVL





patens

ASISLEYRRECHSTDVVESLTNSKMDIQGNDSDPSRPCEYVHLLRKQDSSNQEILR




(Moss)
GMTKW






A0A0B9G

Photo-

MMRFLSVIFFLVFTQHALAAKLMVLGDSLSAGYQMQAEQSWPNLLDAELEKYG
161


JL1

bacterium

HEVTVVNASISGDTTGNGMARLPRLLEQHQPDFVLIELGANDGLRGFPPTTIRNNL





gaetbulicola

GEMITQIEQAGAYPLLMQIVVPPNYGKRYSDQFAKVYQEISNTLDIPLLPFFLEHII





LKQEWMMEDGLHPKPDAQPWIANFMANEIAPHL






Q9LK77

Arabidopsis

MNSPRPISVVSTFASPSSTSDPIRKPLSLWPGMYHSPVTTALWEARSKIFESLLDPP
162




thaliana

KDAPPQSQLLTRTPSHSRTTIFYPFSTDFILREQYRDPWNEVRIGILLEDLDALAGTI




(Mouse-
SVKHCSDDDSTTRPLLLVTASVHKIVLKKPICVDIDLKIVASVIWVGRSSIEIQLEV




ear cress)
MQSELKDVKASSDSVALTANFIFVARDSKTGKAAPINRLSPETEVEKLLFEEAEAR





NNLRKKKRGGDRREFDHGECKKLEAWLAEGRIFSDMPALADRNSILLKDTRLEN





SLICQPQQRNIHGRIFGGFLMHRAFELAFSTAYTFAGLVPYFLEVDHVDFLRPVDV





GDFLRFKSCVLYTQLDKQDCPLINIEVVAHVTSPEIRSSEVSNTFYFKFTVRPEAKA





RNNGFKLRNVVPATEEEARHILERMDAEALKSSKQQCVGTILQ






Q01FC4

Ostreococcus

MIARASGASDVASADRSVAKPTANGEKSFSGMDGTEWFSRNFSEQGRKFSEVFP
163




tauri

VRYAETGPNGEATMVTIADLIQECACNHAQGIWGVGQSMPAEMAKGHLAWVCT





RLHLCVRKYPKWGEKMEVSTWFEPQGKIAARRDYSITDESGVQIGEATSQWVVL





NLNTRRMARIPNSVLEDFKYQALERQVMEEGYASDKLADVTEIAANQCVSPITH





HVRRNDMDMNGHVNNVVYVQWILESVPQETWNGRALQEIILEYRSECNFGECIT





ATCCEVEEQSDSYVLLHKLARGDDEIVRAKTVWTKQKTS






P0AGG2

Escherichia

MSQALKNLLTLLNLEKIEEGLFRGQSEDLGLRQVFGGQVVGQALYAAKETVPEE
164




coli

RLVHSFHSYFLRPGDSKKPIIYDVETLRDGNSFSARRVAAIQNGKPIFYMTASFQA




(strain
PEAGFEHQKTMPSAPAPDGLPSETQIAQSLAHLLPPVLKDKFICDRPLEVRPVEFH




K12)
NPLKGHVAEPHRQVWIRANGSVPDDLRVHQYLLGYASDLNFLPVALQPHGIGFL





EPGIQIATIDHSMWFHRPFNLNEWLLYSVESTSASSARGFVRGEFYTQDGVLVAST





VQEGVMRNHN






Q3B7M2

Bos

MVLGRGLLGRWSVAELGAVCARLGLGPALLGSLHHLGLRKSLTVDQGTMKVEL
165




taurus

LPALTDNYMYLLIDEDTKEAAIVDPVQPQKVVETARKHGVKLTTVLTTHHHWD




(Bovine)
HAGGNEKLVKLEPGLKVYGGDDRIGALTHKVTHLSTLQVGSLHVKCLSTPCHTS





GHICYFVTKPNSPEPPAVFTGDTLFVAGCGKFYEGTADEMYKALLEVLGRLPADT





RVYCGHEYTINNLKFARHVEPDNTAVREKLAWAKEKYSIGEPTVPSTIAEEFTYN





PFMRVREKTVQQHAGETEPVATMRAIRKEKDQFKMPRD






I3S4A5

Aledicago

MPSWFDIHEIPVTANSPNDESSLLKAVQNVHATIDKEIAAGTNPNNIFICGFSQGG
166




truncatula

ALTLASVLLYPKTLGGGAVFSGWVPFNSSVIEQITPEAKRTPILWSHGLSDKTVLF




(Barrel
EARQAAPPFLEKIGVSCEFKAYPGLAHSINNEELKHLESWIKARLQSSS




medic)





(Nledicago






tribuloides)








E4MWI3

Eutrema

MESAMNTESVFEFLGNVPLLQKLPSSSLKKIAQVVVLKRYGKGDYVIREDQAWD
167




halophilum

GCYFIFAGEAQVSGPAEEENRSEFLLKKYDYFGHGISAHVHSADIIATSELTCLVLP




(Salt
RDHCRLLETNSIWQSDKEVQKCSLVERILHLDPLELNIFRGITLPDAPKFGKVFGG




cress)
QFMGQALAAASKTVDFLKIVHSLHSYFLLVGDIDIPIIYQVHRIRDGNNFATRRVD




(Sisymbrium
AIQKGNIIFILLASFQKEQQGFDHQESTMPSAPDPDTLLSLEELRERRITDPHLPRSY





halophilum)

RNKVATANFVPWPIDIRFCDPSNSTNQTKSPPRLRYWFRAKGKLSDDQALHRCV





VAFASDLIFASVSLNPHRRKGLRSAALSLDHAMWFHRPLRADDWLLFVIVSPTAH





MTRGFVSGQMFNRKGELVVSLTQEALLREARPPKPSVTSKL






A9NV70

Picea

MYHSPVTNALWHARSSIFERLLDPSVDAPPQSQLLSKTPSQSRTSILYNFSSDYILR
168




sitchensis

EQYRDPWNEVRIGKLLEDLDALAGTIAVKHCSDDDSTTRPLLLVTASVDKMVLK




(Sitka
KPIRVDTDLKVAGAVTWVGRSSLEIQMVITQPPEGETETGDSVALTANFMFVARD




spruce)
SKTGKSALINRLLPQTEQEKALLAEGEARDMRRKKERQRQGKEFEEGHRLHGDG




(Pinus
DRLKALLREGRVLCDMPALADRDSMLIKDTRLENALICQPQQRNLHGRIFGGFL





sitchensis)

MHRASELAFSTCYAFVGHTPLFLEVDHVDFLRPVDVGDFLRFKSCVLFTQVDDPK





RPLIDIEVVAHVIRPELRSSEVSNTFYFTFTVHPVALEGGLKIRKVLPATEEEARHV





LERIDAENLN






M1Z1V0
[Clostridium]
MKATPLYIKDYKVEINHVDFKGDLKLSSLFTYCQDIAGLHAENLGMGREVLYTQ
169




ultunense

HRVIWVLVRVRVDIIKYPKWKDILTLETWPQEPSRMGFDRDFLIKDKKGNILAKA




Esp
VSTWVVIDVESRKLVRTKSVYTGYPLVVEKRAIDCKLGNLKSSGELETAYERTVR





YSDIDVNEHLNNAKYLDFIMDSFSFEEHRRFNVKSVEISYSNEALLGETIKIYEDRS





RIDSNIIYMEGIREGKDLVFKSQIEIEEK






R6XLC3

Clostridium

MAIIENKYHIGIKYVDKDRLLSLRGIILLFEDIACRHSDMVGYGINDVTKTHFSWV
170



sp.
LLNWKIKVLSRINYGSIVTVKTWSRETSKLYTYRDFEIYDENNNLICIASSKWVLL




CAG: 798
STETGHIIHITEEIKNAYLAENKTVFNESDLHKIVEPDNAEKTFSFTVRRRDIDINNH





MNNLYYLDYALEALPEEVYSKFFNNVEIMYKYSAKLGETINCFYKEEEDGYYVM





MKSATDNILHALVKLY






R5TEH3

Roseburia

MYQFKSRVRFSEVDSQLHMTLPSIINYFQDCSTFHSDSIGYGIEVMMEQGYAWILS
171



sp.
SWQIIINRYPKFGEEITVSTWAHGWKAFFGYRNFKLEDTEGNLLAYANTNWIYM




CAG: 50
NIRTGHPERIPKEICDLYKCEPALPMEESSRKITPPAKGTGITPIQVHRYDIDSNNHV





NNERYVPMAMECLPEGAQIRQLRVEYRNSAVYGDTIYPVYHQEEDLLKVSLNDS





DGKPYAIVEFQLAPLSDQAD






E6MF99

Pseudora

MGKIFERPQAIATYDCLEDHHLSPVAVMNYFQQISLEHSASLKAGPYELSALDLT
172




mibacter

WIVVKYHVDFWQMPRFLDQLQLGTWASAFKGFTAHRGFFLKNQSGEHMVDGQ





alactolyticus

SHWMMVDRRQNHIVRVNEVPINAVYDVEDQGPRFKMPRLARIKDWENVRQFSV




ATCC
RYLDIDYNGHVNNVCYLAWALACLPAVVLQTRTLKTLDIVFKEQALYGDVVTV




23263
KDREIAPNCYRVDIFNANETLLTQLQLQF









In some embodiments, the amino acid corresponding to position 5 is substituted with tyrosine (Y) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 35 is substituted with serine (S) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 38 is substituted with glutamine (Q) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 64 is substituted with valine (V) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 241 is substituted with glutamic acid (E) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 45 is substituted with methionine or isoleucine (I) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 128 is substituted with tyrosine (Y) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 175 is substituted with serine (S) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 33 is substituted with aspartic acid (D) or an equivalent amino acid, and the amino acid corresponding to position 128 is substituted with tyrosine (Y) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 59 is substituted with valine (V) or an equivalent amino acid, and the amino acid corresponding to position 90 is substituted with phenylalanine (F) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 40 is substituted with glutamic acid (E) or an equivalent amino acid, and the amino acid corresponding to position 111 is substituted with tryptophan (W) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 36 is substituted with glycine (G) or an equivalent amino acid, and the amino acid corresponding to position 128 is substituted with tyrosine (Y) or an equivalent amino acid. In some embodiments, the amino acid corresponding to position 32 is substituted with glutamine (Q) or an equivalent amino acid, and the amino acid corresponding to position 40 is substituted with glutamic acid (E) or an equivalent amino acid.


In some embodiments substitution of one or more amino acid(s) corresponding to position(s) 5, 35, 38, 64 and 241, as described above, increases enzymatic activity and/or improves substrate specificity. In some embodiments, substitution of one or more amino acid(s) in the substrate binding site, for example, one or more amino acid(s) corresponding to position(s) 45, 128, and 175, as described above, increases enzymatic activity and/or improves substrate specificity. In some embodiments, substitution of pairs of amino acid residues at the substrate binding site, for example, one or more amino acid(s) corresponding to positions 33 and 128; positions 59 and 90; positions 40 and 111; positions 36 and 128; positions 32 and 40; as described above, increases enzymatic activity and/or improves substrate specificity.


It is understood that any wild-type acyl-ACP TE can be mutated with any combination of the amino acid substitutions described to obtain a polypeptide with increase enzymatic activity and/or improved substrate specificity. In some embodiments, the mutant acyl-ACP TE comprises two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more or ten or more substitutions at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1.


As set forth above, the mutant acyl-ACP TE can have one or more amino acid substitutions described above, relative to a wild-type acyl-ACP TE, for example, and not to be limiting, relative to a wild-type acyl-ACP TE classified under EC 3.1.2.- or EC 3.1.2.14. In some embodiments, the mutant acyl-ACP TE has one or more additional modifications. Protein modifications include amino acid sequence modifications. Modifications in amino acid sequence may arise naturally as allelic variations (e.g., due to genetic polymorphism), may arise due to environmental influence (e.g., by exposure to ultraviolet light), or may be produced by human intervention (e.g., by mutagenesis of cloned DNA sequences), such as induced point, deletion, insertion, and substitution mutants. Modifications in a nucleic acid encoding an acyl-ACP TE can result in changes in the amino acid sequence, provide silent mutations, modify a restriction site, or provide other specific mutations. Amino acid sequence modifications typically fall into one or more of three classes: substitutional, insertional, or deletional modifications. Insertions include amino and/or terminal fusions as well as intrasequence insertions of single or multiple amino acid residues. Insertions ordinarily will be smaller insertions than those of amino or carboxyl terminal fusions, for example, on the order of one to four residues. Deletions are characterized by the removal of one or more amino acid residues from the protein sequence. Typically, no more than about from 2 to 6 residues are deleted at any one site within the protein molecule. Amino acid substitutions are typically of single residues, but can occur at a number of different locations at once; insertions usually will be on the order of about 1 to 10 amino acid residues; and deletions will range from about 1 to 30 residues. Deletions or insertions preferably are made in adjacent pairs, i.e., a deletion of 2 residues or insertion of 2 residues. Substitutions, deletions, insertions or any combination thereof may be combined to arrive at a final construct. Substitutional modifications are those in which at least one residue has been removed and a different residues inserted in its place. In some embodiments, conservative or equivalent substitutions are made. A conservative substitution results in substitution of an amino acid with a chemically and/or functionally similar amino acid. Modifications, including specific amino acid substitutions, are made by known methods.


By way of example, modifications are made by site-specific mutagenesis of nucleotides in the DNA encoding the protein, thereby producing DNA encoding the modification, and thereafter expressing the DNA in recombinant cell culture. Techniques for making substitution mutations at predetermined sites in DNA having a known sequence are well known, for example, PCR mutagenesis, primer extension or inverse PCR mutagenesis.


Fusion polypeptides comprising any of the polypeptides described herein are also provided. The polypeptides can be fused to heterologous sequences, for example, and not to be limiting, tags or sequences designed to facilitate expression, purification and/or detection of recombinantly-expressed proteins. Non-limiting examples include a periplasmic tag, a poly-histidine tag, a maltose binding protein (MBP), Protein A, glutathione S-transferase (GST), fluorescent protein sequences (e.g. GFP), and epitope tags such as myc, FLAG, and haemagglutinin tags.


In some embodiments, the amino acid sequence of the polypeptide having an acyl-ACP TE activity, for example, a mutant acyl-ACP TE has at least 50%, 55% 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% amino acid sequence identity with the amino acid sequence of the wild-type acyl-ACP TE, and has enzymatic activity. In certain embodiments, the amino acid sequence of the polypeptide having an acyl-ACP TE activity, for example, a mutant acyl-ACP TE has at least 90%, 95%, 98%, 98.1, 98.2%, 98.3%, 98.4%, 98.5%, 98.6%, 98.7%, 98.8%, 98.9%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9% sequence identity to the wild-type acyl-ACP TE and has enzymatic activity. In certain embodiments, the enzymatic activity is specific for pimeloyl-ACP.


Also provided herein are fragments of the polypeptides described herein. These fragments preferably include at least 20 contiguous amino acids, more preferably at least 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 or more contiguous amino acids. In some embodiments, the polypeptide comprises a fragment of a mutant acyl-ACP TE described herein.


Derivatives of any of the mutant polypeptides described herein also provided. In some embodiments, derivative polypeptides are polypeptides that have been further altered, for example by conjugation or complexing with other chemical moieties, by post-translational modification (e.g. phosphorylation, acetylation and the like), modification of glycosylation (e.g. adding, removing or altering glycosylation), and/or inclusion/substitution of additional amino acid sequences as would be understood in the art. Derivatives also include fusion proteins, as described above. Other derivatives contemplated by the embodiments include, but are not limited to, modification to side chains, incorporation of unnatural amino acids and/or their derivatives during peptide, or protein synthesis and the use of crosslinkers and other methods which impose conformational constraints on the polypeptides and fragments thereof.


Nucleic Acids and Vectors

In another embodiment, a nucleic acid encoding any of the polypeptides having an acyl-ACP TE activity described herein is provided. In some embodiments, the nucleic acid encodes a polypeptide having an acyl-ACP TE activity, wherein the polypeptide comprises one or more amino acid substitution(s) relative to a wild-type acyl-ACP TE, wherein the one or more amino acid substitution(s) are at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1. In some embodiments, the nucleic acid encodes a polypeptide comprising a mutant acyl-ACP TE or a functional fragment thereof. In some embodiments, the nucleic acid encodes a polypeptide comprising a mutant acyl-ACP TE, wherein the mutant acyl-ACP TE comprises one or more amino acid substitution(s) relative to a wild-type acyl-ACP TE, wherein the one or more amino acid substitution(s) are at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1, or a functional fragment thereof. In some embodiments, the enzyme is an acyl-ACP TE classified under EC 3.1.2.-. In some embodiments, the wild-type acyl-ACP TE is classified under EC 3.1.2.14. Vectors or genetic constructs, including expression vectors, comprising any of the nucleic acid sequences disclosed herein are also provided.


In some embodiments, the nucleic acid encoding the polypeptide having an acyl-ACP-TE activity, for example, a mutant acyl-ACP TE has at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% sequence identity with the nucleic acid sequence encoding the wild-type acyl-ACP TE. In certain embodiments, the nucleic acid sequence encoding the polypeptide having an acyl-ACP-TE activity, for example, a mutant acyl-ACP TE, has at least 90%, 95%, 98%, 98.1, 98.2%, 98.3%, 98.4%, 98.5%, 98.6%, 98.7%, 98.8%, 98.9%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9% sequence identity to the nucleic acid encoding a wild-type acyl-ACP TE.


Nucleic acid molecules that are fragments of these nucleic acid sequences encoding polypeptides are also encompassed by the embodiments. By “fragment” is intended a portion of the nucleic acid sequence encoding a portion of a polypeptide. In some embodiments, the nucleic acid has been codon optimized for expression of any one of the polypeptides described herein.


Some embodiments relate to nucleic acids consisting essentially of or consisting of a nucleic acid sequence encoding a polypeptide as described above and elsewhere herein. In some embodiments, the nucleic acid sequence is a DNA sequence, for example, a cDNA sequence. In other embodiments, the nucleic acid sequence is an RNA sequence. In some embodiments, the nucleic acid is a cDNA encoding any of the polypeptides described herein. The nucleotide sequences encoding the polypeptide may be prepared by any suitable technologies well known to those skilled in the art, including, but not limited to, recombinant DNA technology and chemical synthesis. Synthetic polynucleotides may be prepared using commercially available automated polynucleotide synthesizers.


In some embodiments, the polynucleotide comprises a sequence encoding any one of the polypeptides described herein operably linked to a sequence encoding another protein, which can be a fusion protein or another protein separated by a linker. In some embodiments, the linker has a protease cleavage site, such as a cleavage site for Factor Xa or thrombin, which allow the relevant protease to partially digest the fusion polypeptide described herein and thereby liberate the recombinant polypeptide therefrom. The liberated polypeptide can then be isolated from the fusion partner by, for example, subsequent chromatographic separation.


Some embodiments provide genetic constructs in the form of, or comprising genetic components of, a plasmid, bacteriophage, a cosmid, a yeast or bacterial artificial chromosome, as are well understood in the art. Genetic constructs may be suitable for maintenance and propagation of the isolated nucleic acid in bacteria or other host cells, for manipulation by recombinant DNA technology and/or expression (expression vectors) of the nucleic acid or an encoded polypeptide as described herein.


Some embodiments relate to recombinant expression vectors comprising a DNA sequence encoding one or more of the polypeptides described herein. In some embodiments, the expression vector comprises one or more of said DNA sequences operably linked to a promoter. Suitably, the expression vector comprises the nucleic acid encoding one of the polypeptides described herein operably linked to one or more additional sequences. In some embodiments, the expression vector may be either a self-replicating extra-chromosomal vector such as a plasmid, or a vector that integrates into a host genome. Non-limiting examples of viral expression vectors include adenovirus vectors, adeno-associated virus vectors, herpes viral vectors, retroviral vectors, lentiviral vectors, and the like. For example, adenovirus vectors can be first, second, third, and/or fourth generation adenoviral vectors or gutless adenoviral vectors. Adenovirus vectors can be generated to very high titers of infectious particles, infect a great variety of cells, efficiently transfer genes to cells that are not dividing, and are seldom integrated in the host genome, which avoids the risk of cellular transformation by insertional mutagenesis. The vector may further include sequences flanking the polynucleotide giving rise to RNA which comprise sequences homologous to eukaryotic genomic sequences or viral genomic sequences. This will allow the introduction of the polynucleotides described herein into the genome of a host cell.


An integrative cloning vector may integrate at random or at a predetermined target locus in the chromosome(s) of the host cell into which it is to be integrated.


Expression of the Polypeptides

Any of the polypeptides disclosed herein, including mutant or variant polypeptides, can be expressed using conventional methods known to those of skill in the art. Generally, for production of acyl-ACP TEs in E. coli the T7 expression system can be used. Genes for acyl-ACP TEs can be cloned into a vector under the control of the T7 promoter and transformed into a BL21[DE3] E. coli host. Expression can then be induced by the addition of 1 mM IPTG to the culture media. In some embodiments, a nucleic acid sequence encoding any of the polypeptides disclosed herein, for example, a DNA sequence, can be expressed, using an expression vector, which typically includes control sequences encoding a promoter, operator, ribosome binding site, translation initiation signal, and, optionally, a repressor gene or various activator genes. In some embodiments, the expression vector optionally comprises a suitable transcription terminator. In some embodiments, polyadenylation sequences operably connected to the DNA sequence encoding the polypeptide are used for expression in eukaryotic cells. Termination and polyadenylation sequences may suitably be derived from the same sources as the promoter or not. One of skill in the art would know which combination of an expression vector, including specific elements of the vector, can be used with one or more host cells to produce the desired polypeptide. In some embodiments, the vector comprises a selectable marker. Selectable markers are well known in the art and will vary with the host cell used. Suitable selection markers can include, for example, genes coding for ampicillin and/or tetracycline resistance, which enable cells transformed with these vectors to grow in the presence of these antibiotics.


In some embodiments, a host cell comprising a vector described herein is cultured. After reaching a desired cell density or titer of the polypeptide, the culture is stopped and the polypeptide is recovered using known procedures. Alternatively, the host cell is used directly (e.g., pellet, suspension), i.e., without isolation of the recombinant protein.


The recombinant expression vector carrying the DNA sequence encoding a polypeptide as described herein may be any vector, which may conveniently be subjected to recombinant DNA procedures, and the choice of vector will often depend on the host cell into which it is to be introduced. Thus, the vector may be an autonomously replicating vector, i.e., a vector that exists as an extrachromosomal entity, e.g., a plasmid, a bacteriophage or an extrachromosomal element, minichromosome, or an artificial chromosome, the replication of which is independent of chromosomal replication. Alternatively, 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.


In the vector, the DNA sequence typically is operably connected to a suitable promoter sequence. The promoter may be any DNA sequence that shows transcriptional activity in the host cell of choice and may be derived from genes encoding proteins either homologous or heterologous to the host cell. Examples of suitable promoters for directing the transcription of the DNA sequence encoding a polypeptide as described herein, especially in a bacterial host, are the promoter of the lac operon of E. coli, the Streptomyces coelicolor agarase gene dagA promoters, the promoters of the Castellaniella defragrans, and others. For transcription in a fungal host, examples of useful promoters are those derived from the gene encoding A. oryzae TAKA amylase, Rhizomucor miehei aspartic proteinase, A. niger neutral LDH, A. niger acid stable LDH, A. niger glucoamylase, Rhizomucor miehei lipase, A. oryzae alkaline protease, A. oryzae triose phosphate isomerase or A. nidulans acetamidase. The promoters can be selected based on the desired outcome. The nucleic acids can be combined with constitutive, tissue-preferred, inducible, or other promoters for expression in the host cell or organism. The above list of promoters is not meant to be limiting. Any appropriate promoter can be used in the embodiments.


In some embodiments, the expression vector described may also comprise a suitable transcription terminator and, in eukaryotes, polyadenylation sequences operably connected to the DNA sequence encoding the polypeptide as described herein. Termination and polyadenylation sequences may suitably be derived from the same sources as the promoter or not.


In some embodiments, the vector may further comprise a DNA sequence enabling the vector to replicate in the host cell in question. Examples of such sequences are the origins of replication of plasmids pUC19, pACYC177, pUB110, pE194, pAMB1 and pIJ702. The above list of origins of replication is not meant to be limiting. Any appropriate origins of replication can be used in the embodiments


In some embodiments, the vector may also comprise a selectable marker. Selectable marker genes are utilized for the selection of transformed cells or tissues, e.g., a gene the product of which complements a defect in the host cell, such as the dal genes from B. subtilis or B. licheniformis, or one which confers antibiotic resistance such as ampicillin, kanamycin, chloramphenicol or tetracyclin resistance. Furthermore, the vector may comprise Aspergillus selection markers such as amdS, argB, niaD and sC, a marker giving rise to hygromycin resistance, or the selection may be accomplished by co-transformation. The above list of selectable marker genes is not meant to be limiting. Any selectable marker gene can be used in the embodiments.


Appropriate culture mediums and conditions for the above-described host cells are known in the art. While intracellular expression may be advantageous in some respects, e.g., when using certain bacteria as host cells, it might be preferred that the expression is extracellular or periplasmic.


Enzymatic Activity and Substrate Specificity

In some embodiments, any of the polypeptides described herein has increased enzymatic activity and/or improved substrate specificity as compared to a wild-type acyl-ACP TE. In some embodiments, any of the polypeptides described herein comprises, consists essentially of or consists of a mutant acyl-ACP that has increased enzymatic activity and/or improved substrate specificity relative to the wild-type acyl-ACP TE. In some embodiments, any of the polypeptide described herein comprises, consists essentially of or consists of a mutant acyl-ACP that has increased enzymatic activity relative to the wild-type acyl-ACP TE. In some embodiments, any of the polypeptides described herein comprises, consists essentially of or consists of a mutant acyl-ACP TE that has improved substrate specificity relative to the wild-type acyl-ACP TE. In some embodiments, any of the polypeptides described herein comprises, consists essentially of or consists of a mutant acyl-ACP that has increased enzymatic activity and/or improved substrate specificity for pimeloyl-ACP relative to the wild-type acyl-ACP TE.


In some embodiments, a polypeptide, for example, a polypeptide comprising a mutant acyl-ACP TE has an increase in enzymatic activity of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 300%, 400%, or at least 500% relative to the activity of the wild-type acyl-ACP TE. In certain embodiments, enzymatic activity is specific activity, i.e., enzymatic activity for reaction of the mutant acyl-ACP TE with a specific substrate, for example, pimeloyl-ACP. In some embodiments, the increase in enzymatic activity results in an increase in the production of 7-AHA when the polypeptide is expressed in a host cell or organism, as described herein.


In some embodiments, the improved substrate specificity of the polypeptide, for example, a mutant acyl-ACP TE, relative to the substrate specificity of the wild-type acyl-ACP TE results in a polypeptide that can convert at least one substrate into at least one product at a kcat (s−1) greater than that of the wild-type acyl-ACP TE.


In some embodiments, the improved substrate specificity of the polypeptide, for example, a mutant acyl-ACP TE, relative to the activity of the wild-type acyl-ACP TE results in a polypeptide that can convert at least one substrate into at least one product at a Km lower than that of the wild-type acyl-ACP TE. In some embodiments, the improved substrate specificity is improved substrate specificity for pimeloyl-ACP.


Host Cells and Organisms

Any of the nucleic acids disclosed herein can be stably or transiently introduced into an organism, such as a host cell, using established techniques, including, but not limited to, electroporation, calcium phosphate precipitation, liposome-mediated transfection, contact with nanowires or nanotubes, spheroplasting, PEG 1000-mediated transformation, biolistics, lithium acetate transformation, lithium chloride transformation, and the like.


For stable transformation, a subject nucleic acid will generally further include a selectable marker, e.g., any of several well-known selectable markers such as neomycin resistance, and the like. Some embodiments relate to host cells comprising an exogenous DNA molecule (i.e., a molecule not otherwise present in the wild-type counterpart cell) encoding a polypeptide as described herein. In some embodiments, these host cells can be described as expression systems. Suitable host cells for expression may be prokaryotic or eukaryotic. Without limitation, suitable host cells may be mammalian cells (e.g. HeLa, HEK293T, Jurkat cells), yeast cells (e.g. Saccharomyces cerevisiae), insect cells (e.g. Sf9, Trichoplusia ni) used with or without a baculovirus expression system, or bacterial cells, such as E. coli (Origami2(DE3), BL21(DE3)), or a vaccinia virus host. Introduction of genetic constructs into host cells (whether prokaryotic or eukaryotic) is well known in the art, for example, as described in Current Protocols in Molecular Biology Eds. Ausubel et al., (John Wiley & Sons, Inc. current update Jul. 2, 2014).


A further embodiment relates to a transformed or transduced organism or microorganism (i.e., an engineered organism), such as an organism selected from plant cells, insect cells, bacteria, yeast, baculovirus, protozoa, nematodes, algae, and transgenic mammals (e.g., mice, rats, pigs). The microorganisms include prokaryotic and eukaryotic microbial species from the domains Archaea, Bacteria and Eucarya, the latter including yeast and filamentous fungi, protozoa, algae, or higher Protista. The terms “microbial cells” and “microbes” are used interchangeably with the term microorganism. The transformed organism comprises a DNA molecule of the embodiments, an expression cassette comprising the DNA molecule or a vector comprising the expression cassette of the disclosed embodiments, which may be stably incorporated or not stably incorporated into the genome of the transformed organism. Other suitable organisms also include synthetic cells or cells produced by synthetic genomes as described in Venter et al. US Pat. Pub. No. 2007/0264688, and cell-like systems or synthetic cells as described in Glass et al. US Pat. Pub. No. 2007/0269862.


In certain embodiments, the host is a prokaryote selected from the group consisting of Escherichia; Clostridia; Corynebacteria; Cupriavidus; Pseudomonas; Delftia; Bacillus; Lactobacillus; Lactococcus; and Rhodococcus, or a eukaryote selected from the group consisting of Aspergillus, Saccharomyces, Pichia, Yarrowia, Issatchenkia, Debaryomyces, Arxula, and Kluyveromyces.


In some embodiments, the host microorganism is a prokaryote. For example, the prokaryote can be from the bacterial genus Escherichia such as Escherichia coli; from the bacterial genus Clostridia, such as Clostridium ljungdahlii, Clostridium autoethanogenum or Clostridium kluyveri; from the bacterial genus Corynebacteria, such as Corynebacterium glutamicum; from the bacterial genus Cupriavidus, such as Cupriavidus necator or Cupriavidus metallidurans; from the bacterial genus Pseudomonas, such as Pseudomonas fluorescens, Pseudomonas putida or Pseudomonas oleavorans; from the bacterial genus Delftia such as Delftia acidovorans; from the bacterial genus Bacillus such as Bacillus subtillis; from the bacterial genus Lactobacillus, such as Lactobacillus delbrueckii; or from the bacterial genus Lactococcus, such as Lactococcus lactis.


In some embodiments, the host microorganism is a eukaryote (e.g., a fungus such as a yeast). For example, the eukaryote can be from the fungus genus Aspergillus such as Aspergillus niger; from the yeast genus Saccharomyces, such as Saccharomyces cerevisiae; from the yeast genus Pichia such as Pichia pastoris; from the yeast genus Yarrowia such as Yarrowia hpolytica; from the yeast genus Issatchenkia, such as Issathenkia orientalis; from the yeast genus Debaryomyces such as Debaryomyces hansenii; from the yeast genus Arxula such as Arxula adenoinivorans; or from the yeast genus Kluyveromyces such as Kluyveromyces lactis.


Exemplary species of yeast or fungi species include any species selected from the order Saccharomycetales, family Saccaromycetaceae, including the genera Saccharomyces, Kluyveromyces and Pichia; the order Saccharomycetales, family Dipodascaceae, including the genus Yarrowia; the order Schizosaccharomycetales, family Schizosaccaromycetaceae, including the genus Schizosaccharomyces; the order Eurotiales, family Trichocomaceae, including the genus Aspergillus; and the order Mucorales, family Mucoraceae, including the genus Rhizopus. Non-limiting species of host yeast or fungi include Saccharomyces cerevisiae, Schizosaccharomyces pombe, Kluyveromyces lactis, Kluyveromyces marxianus, Aspergillus terreus, Aspergillus niger, Pichia pastoris, Rhizopus arrhizus, Rhizopus oryzae, Yarrowia lipolytica, and the like. E. coli is a particularly useful host organism since it is a well characterized microbial organism suitable for genetic engineering. Other particularly useful host organisms include yeast such as Saccharomyces cerevisiae. It is understood that any suitable microbial host organism can be used to introduce metabolic and/or genetic modifications to produce a desired product.


Another embodiment of the disclosure comprises a method of making a polypeptide from a recombinant nucleic acid sequence disclosed herein comprising: a) providing a population of host cells or organisms; and b) growing the population of cells or organisms under conditions whereby the polypeptide encoded by the nucleic acid of the disclosure is expressed; and c) isolating the resulting polypeptide.


The host cells can be fermented to produce a polypeptide having acyl-ACP TE activity described herein or to catalyze the reactions that are catalyzed by the polypeptides described herein. Fermentation broadly refers to the conversion of organic materials into target substances by host cells, for example, the conversion of a carbon source by recombinant host cells into fatty acids or derivatives thereof by propagating a culture of the recombinant host cells in a media comprising the carbon source. As used herein, conditions permissive for the production means any conditions that allow a host cell to produce a desired product, such as a fatty acid or a fatty acid derivative. Similarly, conditions in which the polynucleotide sequence of a vector is expressed means any conditions that allow a host cell to synthesize a polypeptide. Suitable conditions include, for example, fermentation conditions. Fermentation conditions can comprise many parameters, including but not limited to temperature ranges, levels of aeration, feed rates and media composition. Each of these conditions, individually and in combination, allows the host cell to grow. Fermentation can be aerobic, anaerobic, or variations thereof (such as micro-aerobic). Exemplary culture media include broths or gels. Generally, the medium includes a carbon source that can be metabolized by a host cell directly. In addition, enzymes can be used in the medium to facilitate the mobilization (e.g., the depolymerization of starch or cellulose to fermentable sugars) and subsequent metabolism of the carbon source. For small scale production, the engineered host cells can be grown in batches of, for example, about 100 mL, 500 mL, 1 L, 2 L, 5 L, or 10 L; fermented; and induced to express a desired polynucleotide sequence, such as a polynucleotide sequence encoding a polypeptide comprising a mutant acyl-ACP TE disclosed herein. For large scale production, the engineered host cells can be grown in batches of about 10 L, 100 L, 1000 L, 10,000 L, 100,000 L, 1,000,000 L or larger; fermented; and induced to express a desired polynucleotide sequence. Alternatively, large scale fed-batch fermentation may be carried out.


Methods

The polypeptides, nucleotides, and cells/organisms described herein are useful for many different applications.


Provided herein, in one embodiment, is a method for producing pimelic acid, pimelate semialdehyde or 7-AHA or any product of FIG. 2, comprising the step of enzymatically converting pimeloyl-ACP to pimelic acid in the presence of any one of the polypeptides described herein.


In a related embodiment, also provided is a method for producing pimelic acid, pimelate semialdehyde or 7-AHA or any product of FIG. 2, comprising the steps of culturing a host cell comprising a nucleic acid sequence encoding a mutant polypeptide of the disclosure in a suitable medium and recovering the pimelic acid, pimelate semialdehyde or 7-AHA. In some embodiments, the host cell can further comprise a nucleic acid encoding an enzyme that catalyzes a different reaction in a biosynthetic pathway for 7-AHA, for example, an enzyme that catalyzes a reaction in the pathway for the production of 7-AHA, shown in FIG. 2. Exemplary recombinant enzymes that can be used include, but are not limited to, a pimeloyl-ACP methyl ester esterase (bioH) that catalyzes the conversion of pimeloyl-ACP methyl ester to pimeloyl-ACP, a carboxlic acid reductase (CAR) that catalyzes the conversion of pimelic acid to pimelate semialdehyde and a transaminase that converts pimelate semialdehyde to 7-AHA.


Another embodiment relates to the use of the polypeptides described herein to produce intermediates, for example, 7-AHA, in the production of synthetic polymers. In some embodiments, the polypeptides described herein are used to produce intermediates in the production of nylon. In certain embodiments, the polypeptides described herein are used to produce intermediates in the production of Nylon 7.


Products

Other embodiments relate to bioderived or fermentation derived products produced by the methods disclosed herein, for example, bioderived pimelic acid, pimelate semialdehyde and 7-amino heptanoate or any product of FIG. 2. In related embodiments, products comprising a chemical produced from any of the bioderived products produced using the methods disclosed herein are also provided. In some embodiments, the product comprises a nylon intermediate, a polyester, a pharmaceutical, a biofuel, a fragrance or a food additive.


Other embodiments include a bio-based or fermentation-derived product, wherein said product comprises: (i) a composition comprising at least one bio-derived, bio-based or fermentation-derived compound, or salts thereof, produced in the presence any of the polypeptides described herein, or produced from the product of a reaction catalyzed by the any of the polypeptides described herein, or any combination thereof; (ii) a bio-derived, bio-based or fermentation-derived polymer comprising the bio-derived, bio-based or fermentation-derived composition or compound of (i), or any combination thereof; (iii) a bio-derived, bio-based or fermentation-derived resin comprising the bio-derived, bio-based or fermentation-derived compound or bio-derived, bio-based or fermentation-derived composition of (i), or any combination thereof, or the bio-derived, bio-based or fermentation-derived polymer of (ii), or any combination thereof; (iv) a substance obtained by molding the bio-derived, bio-based or fermentation-derived polymer of (ii), or the bio-derived, bio-based or fermentation-derived resin of (iii), or any combination thereof; (v) a bio-derived, bio-based or fermentation-derived formulation comprising the bio-derived, bio-based or fermentation-derived composition of (i), bio-derived, bio-based or fermentation-derived compound of (i), bio-derived, bio-based or fermentation-derived polymer of (ii), bio-derived, bio-based or fermentation-derived resin of (iii), or bio-derived, bio-based or fermentation-derived substance of (iv), or any combination thereof, or; (vi) a bio-derived, bio-based or fermentation-derived semi-solid or a non-semi-solid stream, comprising the bio-derived, bio-based or fermentation-derived composition of (i), bio-derived, bio-based or fermentation-derived compound of (i), bio-derived, bio-based or fermentation-derived polymer of (ii), bio-derived, bio-based or fermentation-derived resin of (iii), bio-derived, bio-based or fermentation-derived formulation of (v), or bio-derived, bio-based or fermentation-derived molded substance of (iv), or any combination thereof.


Compositions

Some embodiments relate to compositions comprising one or more disclosed polypeptides or nucleotides, alone or in combination, including in combination with wild-type acyl-ACP TEs. In some embodiments, the composition comprises one or more disclosed polypeptides with improved enzymatic activity and/or substrate specificity. In some embodiments, the composition comprises one or more polypeptides with improved specific activity for the conversion of pimeloyl-ACP to pimelic acid. In some embodiments, the composition comprises pimeloyl-ACP and one or more polypeptides described herein.


In some embodiments the composition is composed of one or more disclosed polypeptides, from (1) commercial suppliers; (2) cloned genes expressing said polypeptides; (3) complex broth (such as that resulting from growth of a microbial strain or any other host cell in media, wherein the strains/host cells secrete the disclosed polypeptides into the media; (4) cell lysates of strains/host cells grown as in (3); and/or (5) any other host cell material expressing the disclosed polypeptide. Different disclosed polypeptides in a composition may be obtained from different sources.


EMBODIMENTS

The following embodiments are contemplated. All combinations of features and embodiment are contemplated.


Embodiment 1

A polypeptide having an acyl-acyl carrier protein (ACP) thioesterase (TE) activity, wherein the polypeptide having acyl-ACP TE activity comprises one or more amino acid substitution(s) relative to a wild-type acyl-ACP TE, wherein the one or more amino acid substitution(s) are at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1, or a functional fragment thereof.


Embodiment 2

An embodiment of embodiment 1, wherein the wild-type acyl-ACP TE is an acyl-ACP TE classified under EC 3.1.2.-.


Embodiment 3

An embodiment of embodiment 1 or 2, wherein the wild-type acyl-ACP TE is an acyl-ACP TE classified under EC 3.1.2.14.


Embodiment 4

An embodiment of any of the embodiments of embodiment 1-3, wherein the amino acid corresponding to position 5 is substituted with tyrosine (Y) or an equivalent amino acid.


Embodiment 5

An embodiment of any of the embodiments of embodiment 1-4, wherein the amino acid at position 35 is substituted with serine (S) or an equivalent amino acid.


Embodiment 6

An embodiment of any of the embodiments of embodiment 1-5, wherein the amino acid at position 38 is substituted with glutamine (Q) or an equivalent amino acid.


Embodiment 7

An embodiment of any of the embodiments of embodiment 1-6, wherein the amino acid at position 64 is substituted with valine (V) or an equivalent amino acid.


Embodiment 8

An embodiment of any of the embodiments of embodiment 1-7, wherein the amino acid at position 241 is substituted with glutamic acid (E) or an equivalent amino acid.


Embodiment 9

An embodiment of any of the embodiments of embodiment 1-8, wherein the amino acid at position 45 is substituted with methionine or isoleucine (I) or an equivalent amino acid.


Embodiment 10

An embodiment of any of the embodiments of embodiment 1-9, wherein the amino acid at position 128 is substituted with tyrosine (Y) or an equivalent amino acid.


Embodiment 11

An embodiment of any of the embodiments of embodiment 1-10, wherein the amino acid at position 175 is substituted with serine (S) or an equivalent amino acid.


Embodiment 12

An embodiment of any of the embodiments of embodiment 1-11, wherein the amino acid at position 33 is substituted with aspartic acid (D) or an equivalent amino acid, and the amino acid at position 128 is substituted with tyrosine (Y) or an equivalent amino acid.


Embodiment 13

An embodiment of any of the embodiments of embodiment 1-11, wherein the amino acid at position 59 is substituted with valine (V) or an equivalent amino acid, and the amino acid at position 90 is substituted with phenylalanine (F) or an equivalent amino acid.


Embodiment 14

An embodiment of any of the embodiments of embodiment 1-11, wherein the amino acid at position 40 is substituted with glutamic acid (E) or an equivalent amino acid, and the amino acid at position 111 is substituted with tryptophan (W) or an equivalent amino acid.


Embodiment 15

An embodiment of any of the embodiments of embodiment 1-11, wherein the amino acid at position 36 is substituted with glycine (G) or an equivalent amino acid and the amino acid at position 128 is substituted with tyrosine (Y) or an equivalent amino acid


Embodiment 16

An embodiment of any of the embodiments of embodiment 1-11, wherein the amino acid at position 32 is substituted with glutamine (Q) or an equivalent amino acid, and the amino acid at position 40 is substituted with glutamic acid (E) or an equivalent amino acid.


Embodiment 17

An embodiment of any of the embodiments of embodiment 1-16, wherein the amino acid sequence of the polypeptide has at least 50% amino acid sequence identity with the amino acid sequence of the wild-type acyl-ACP TE.


Embodiment 18

An embodiment of any of the embodiments of embodiment 1-17, wherein the polypeptide has increased enzymatic activity and/or improved substrate specificity relative to the wild-type acyl-ACP TE.


Embodiment 19

An embodiment of embodiment 18, wherein the polypeptide has improved substrate specificity for pimeloyl-ACP relative to the wild-type acyl-ACP TE.


Embodiment 20

An embodiment of embodiment 18, wherein the polypeptide has an increase in enzymatic activity of at least 10% relative to the wild-type acyl-ACP TE.


Embodiment 21

A nucleic acid sequence encoding the polypeptide of an embodiment of any of the embodiments of embodiment 1-20.


Embodiment 22

A vector or genetic construct comprising the nucleic acid sequence of embodiment 21.


Embodiment 23

An organism or a host cell comprising the vector or genetic construct of embodiment 22, or a functional fragment thereof.


Embodiment 24

The host of embodiment 23, wherein the host is a prokaryotic cell.


Embodiment 25

A composition comprising the polypeptide or functional fragment thereof of an embodiment of any of the embodiments of embodiment 1-20, or the nucleic acid sequence or functional fragment thereof encoding the polypeptide of an embodiment of any of the embodiments of embodiment 1-20, or the vector or genetic construct comprising the nucleic acid sequence of embodiment 21, or the organism or a host comprising the vector or genetic construct of embodiment 22.


Embodiment 26

An embodiment of embodiment 25, further comprising pimeloyl-ACP.


Embodiment 27

A method for producing pimelic acid, pimelate semialdehyde or 7-aminoheptanoate (7-AHA) or any product of FIG. 2, comprising the step of enzymatically converting pimeloyl-ACP to pimelic acid in the presence of any one of the polypeptides or functional fragments thereof of an embodiment of any of the embodiments of embodiment 1-20.


Embodiment 28

A method for producing pimelic acid, pimelate semialdehyde or 7-AHA or any product of FIG. 2, comprising the steps of culturing the host cell of embodiment 23 in a suitable medium and recovering the pimelic acid, pimelate semialdehyde or 7-AHA or other product of FIG. 2.


Embodiment 29

A bio-derived pimelic acid, pimelate semialdehyde or 7-AHA or other product of FIG. 2, that is produced by the method of embodiment 27 or 28.


Embodiment 30

A product comprising a chemical produced from the bio-derived product of embodiment 29, wherein the product comprises a nylon intermediate, a polyester, a pharmaceutical, a biofuel, a fragrance or a food additive.


Embodiment 31

A bio-derived, bio-based or fermentation-derived product, wherein said product comprises: i. a composition comprising at least one bio-derived, bio-based or fermentation-derived compound, or salts thereof, produced in the presence of the polypeptide according to an embodiment of any of the embodiments of embodiment 1-20, or produced from the product of a reaction catalyzed by the polypeptide according to an embodiment of any of the embodiments of embodiment 1-20, or any combination thereof; ii. a bio-derived, bio-based or fermentation-derived polymer comprising the bio-derived, bio-based or fermentation-derived composition or compound of i., or any combination thereof; iii. a bio-derived, bio-based or fermentation-derived resin comprising the bio-derived, bio-based or fermentation-derived compound or bio-derived, bio-based or fermentation-derived composition of i. or any combination thereof or the bio-derived, bio-based or fermentation-derived polymer of ii. or any combination thereof; iv. a substance obtained by molding the bio-derived, bio-based or fermentation-derived polymer of ii. or the bio-derived, bio-based or fermentation-derived resin of iii., or any combination thereof; v. a bio-derived, bio-based or fermentation-derived formulation comprising the bio-derived, bio-based or fermentation-derived composition of i., bio-derived, bio-based or fermentation-derived compound of i., bio-derived, bio-based or fermentation-derived polymer of ii., bio-derived, bio-based or fermentation-derived resin of iii., or bio-derived, bio-based or fermentation-derived substance of iv, or any combination thereof; or vi. a bio-derived, bio-based or fermentation-derived semi-solid or a non-semi-solid stream, comprising the bio-derived, bio-based or fermentation-derived composition of i., bio-derived, bio-based or fermentation-derived compound of i., bio-derived, bio-based or fermentation-derived polymer of ii., bio-derived, bio-based or fermentation-derived resin of iii., bio-derived, bio-based or fermentation-derived formulation of v., or bio-derived, bio-based or fermentation-derived molded substance of iv., or any combination thereof.


While the disclosure has been described in detail, modifications within the spirit and scope of the disclosure will be readily apparent to those of skill in the art. It should be understood that aspects of the disclosure and portions of various embodiments and various features recited above and/or in the appended claims may be combined or interchanged either in whole or in part. In the foregoing descriptions of the various embodiments, those embodiments which refer to another embodiment may be appropriately combined with other embodiments as will be appreciated by one of ordinary skill in the art. Furthermore, those of ordinary skill in the art will appreciate that the foregoing description is by way of example only, and is not intended to limit the disclosure. All US patents and publications cited herein are incorporated by reference in their entirety.


Examples

The following examples are provided by way of illustration only and not by way of limitation. Those of skill in the art will readily recognize a variety of non-critical parameters that could be changed or modified to yield essentially the same or similar results.


Acyl-acyl carrier protein (ACP) thioesterases (TE) play a crucial role in controlling the metabolic flux through the FAS pathway. In particular, acyl-ACP TEs hydrolyze the thioester bonds of acyl-ACPs to release free fatty acids and thereby play essential roles in controlling the amount and composition of fatty acids produced by the fatty acid synthesis (FAS) pathway. This family of enzymes has also received significant interest with regards to metabolic engineering and recently, acyl-ACP TE has attracted even more attention because of increasing interest in producing fatty acids as biofuels or biorenewable chemicals.


Acyl-ACP TEs were previously proposed to utilize a papain-like protease catalytic triad consisting of Cys, His, and Asn. However, the catalytically active Cys residue was not found near the active site of the studied TEs. Crystallographic study of the Oleoyl-ACP-TE (Pdb 2OWN) indicates that residues Asp173, Asn175, His177, and Glu211 play a crucial role for thioesterase activity. These residues correspond to the Asp167, Asn169, His171, and Glu203 in SEQ ID NO: 1 (Uniprot ID No. R5FQ35) one of the top ranked TEs studied. In addition, the Tyr177 was also considered as one of the catalytic residues due to its presence near the proposed catalytic residues. Multiple sequence analyses of the active and inactive Acyl-ACP-TEs indicated that Asp167 and His171 are highly conserved. A three-dimensional model of the Acyl-ACP-TE obtained from Uniport Id No. R5FQ35 shows the catalytic residues (FIG. 3). Analyses also showed that Tyr177 is conserved and substitution of this residue with an aliphatic hydrophobic residue causes substantial loss of enzyme function. Glu203 on the other hand has less of an impact on the catalytic activity of the enzyme. Most surprisingly, the three sequences (Uniprot Nos. B 1ZXQ1, D4KXX4, and H7FRY9) show an activity profile that is restrictive for defining the catalytic residues. This result suggested that, in addition to these residues, other residues could play important roles in catalysis. As described below, the studies described herein identified specific residues and distributions of specific residues in acyl-ACP TEs that can increase enzyme activity and/or affect substrate specificity.


Testing of Thioesterase Enzymes

171 TEs were tested for 7-AHA synthesis in this study. Methods for in vitro and in vivo screening of thioesterase activity are described in U.S. Patent Application Publication No. 20180023103. 54% of the tested enzymes were found to be active. The TE enzymes were ranked based on their activity and subjected to multiple sequence alignment (MSA). The phylogenetic tree constructed from the MSA shows the distribution of the TE over the sequence space. The top 3 TE (from in vitro data) were segregated into 3 clusters. Based on this clustering, it was difficult to identify definite parameters for higher activity. However, the analysis revealed that 63% of the total active TEs are Acyl-ACP-TEs (FIG. 4) that belong to the class of Acyl-ACP hydrolases, or more specifically Oleoyl-ACP TEs (EC 3.1.2.14). This family of enzymes can cleave the thioester bonds from acyl-carrier proteins in the presence of water.


Site specific amino acid distributions were calculated for the active and inactive acyl-ACP-TEs. All acyl-ACP-TEs were subjected to MSA, and distributions of every single residue at each position were calculated. Amino acid population indicated that residue numbers 5, 35, 38, 64, 175, and 241 may play a crucial role in the selectivity and activity of the TEs. The distribution profile indicates a preference for more polar residues at positions 5, 35, and 175 for active enzymes as compared to the inactive enzymes. Interestingly the amino acid at position 241 with a charge in the active enzyme that is the opposite the charge at this position in the inactive enzyme can have significant impact on enzyme activity (FIG. 4A).


Distribution of residues within the substrate binding site showed that three residues can significantly contribute to the activity of the enzyme. Residues Met and Ile at position 45, as well as Tyr and Ser at positions 128 and 175, respectively could play important roles in determining thioesterase activity (FIG. 4B). In addition to the distribution of residues within the binding site, the degree of correlation of the among these positions was calculated.


Statistical coupling analysis (SCA) provides correlation between mutually coupled elements. This method was used to identify the degree of correlation between two residues that are mutually evolved during evolution. See, for example, Shulman et al. “Structural determinants of allosteric ligand activation in RXR heterodimers,” Cell 2004; 116:417-429. In addition, this method provided the statistical distribution of an amino acid for a particular position within the sequence. Given an MSA, the algorithm identifies correlated residues observed within a protein molecule. The SCA method calculates a score for two positions x and y by selecting the most dominant residue at position y and defines a sub-alignment consisting of only sequences having that specific amino acid at position y. The score is calculated using the difference between the amino acids (aa) distribution in full alignment and the subset.





ΔG=GI−GII





Score=Σx=120Σy=120ΔG*F






G
I
=N
x
/N
y (aa fraction at position x in presence of specific aa at y)






G
II
=N
x
/N
(total-MSA) (fraction of aa at position x); F=Observed frequency of each aa pair


To calculate the correlation between substrate binding residues, the acyl-ACP-TEs were divided into three segments: highly active (>100), medium active (>0 and <100), and inactive (below 0). Selecting the top 5 correlated residues from the high, medium, and inactive TEs showed that correlated residues are significantly different for each system (FIG. 5). However, when the correlated residues from highly active enzymes were compared with the other two systems, a better picture was obtained. First of all, these residues were found to have lower correlation in medium active enzymes, and a significant loss in correlation was observed for these residues in the inactive enzymes. In particular, in highly active enzymes, residues Asp33 and Tyr128 are highly correlated, but in the medium active enzymes Tyr128 is changed to Met. Further, these two positions are occupied by two different amino acids (Glu33 and Leu128) in the inactive enzymes (FIG. 5). These results clearly indicate that the residue correlation in the binding site of the enzyme can play an important role in the activity of the enzyme. Considering the potential of the positional correlation for higher activity, the residue pairs shown in the Table 3 could play an important role in increasing enzymatic activity and/or improving substrate specificity of Acyl-ACP TEs.


In summary, residue distribution in the active and inactive TEs suggested that amino acids at positions 5, 35, 38, 64, 175, and 241, along with substrate binding residues at positions 45 and 128 could potentially impact the activity of the Acyl-ACP TEs. Met45, Tyr128, and Ser175 are important residues for substrate specificity and activity. Moreover, the active enzymes were found to have a high degree of correlation among a few residues, which were able to differentiate between the active and the inactive enzymes. Residue pairs Asp33-Tyr128, Val59-Phe90, Glu40-Trp111, Gly36-Tyr128, and Gln32-Glu40 could have an impact on substrate specificity and catalytic activity of the TEs studied for the biosynthesis of 7-AHA.


Therefore, given a wild-type Acyl-ACP-TE, alteration of residues at one or more specific positions could lead to a highly active enzyme for a specific substrate, for example, pimeloyl-ACP.


This is based on the following findings. Higher occurrences of a specific residue at a particular position can have a significant impact on enzyme activity. A specific residue at a given position can also influence the enzymatic activity, for example, increase enzymatic activity. A summary of preferable residues at particular positions is provided in Table 2.












TABLE 2







Position
Preferable residue



















5
Tyr



35
Ser



38
Gln



64
Val



241
Glu










Substrate binding also has a significant preference for residues at particular positions, as provided in Table 3.












TABLE 3







Position
Preferable residue



















45
Met/Ile



128
Tyr



175
Ser










Correlated residues at the substrate binding site were also identified. The pairs of residues provided in Table 4, were found to be highly correlated. This suggests that the presence of one residue at a position can preferentially favor another specific residue at a different site. The top 5 highly correlated residues were considered. For the inactive enzymes, the degree of correlation among these residues were found to be drastically reduced.














TABLE 4







Pos. 1
Res. at Pos. 1
Pos. 2
Res. at Pos. 2





















33
Asp
128
Tyr



59
Val
90
Phe



40
Glu
111
Trp



36
Gly
128
Tyr



32
Gln
40
Glu










Alteration of residues at one or more specific positions can be made using methods for site-directed mutagenesis that are available to those of skill in the art.

Claims
  • 1. A polypeptide having an acyl-acyl carrier protein (ACP) thioesterase (TE) activity, wherein the polypeptide having acyl-ACP TE activity comprises one or more amino acid substitution(s) relative to a wild-type acyl-ACP TE, wherein the one or more amino acid substitution(s) are at amino acid(s) that occupy position(s) corresponding to position(s) 5, 32, 33, 35, 36, 38, 40, 45, 59, 64, 90, 111, 128, 175, and 241 of SEQ ID NO: 1, or a functional fragment thereof.
  • 2. The polypeptide of claim 1, wherein the wild-type acyl-ACP TE is an acyl-ACP TE classified under EC 3.1.2.-.
  • 3. The polypeptide of claim 1, wherein the amino acid corresponding to position 5 is substituted with tyrosine (Y) or an equivalent amino acid.
  • 4. The polypeptide of claim 1, wherein the amino acid at position 35 is substituted with serine (S) or an equivalent amino acid.
  • 5. The polypeptide of claim 1, wherein the amino acid at position 38 is substituted with glutamine (Q) or an equivalent amino acid.
  • 6. The polypeptide of claim 1, wherein the amino acid at position 64 is substituted with valine (V) or an equivalent amino acid.
  • 7. The polypeptide of claim 1, wherein the amino acid at position 241 is substituted with glutamic acid (E) or an equivalent amino acid.
  • 8. The polypeptide of claim 1, wherein the amino acid at position 45 is substituted with methionine or isoleucine (I) or an equivalent amino acid.
  • 9. The polypeptide of claim 1, wherein the amino acid at position 128 is substituted with tyrosine (Y) or an equivalent amino acid.
  • 10. The polypeptide of claim 1, wherein the amino acid at position 175 is substituted with serine (S) or an equivalent amino acid.
  • 11. The polypeptide of claim 1, wherein the amino acid at position 33 is substituted with aspartic acid (D) or an equivalent amino acid, and the amino acid at position 128 is substituted with tyrosine (Y) or an equivalent amino acid.
  • 12. The polypeptide of claim 1, wherein the amino acid at position 59 is substituted with valine (V) or an equivalent amino acid, and the amino acid at position 90 is substituted with phenylalanine (F) or an equivalent amino acid.
  • 13. The polypeptide of claim 1, wherein the amino acid at position 40 is substituted with glutamic acid (E) or an equivalent amino acid, and the amino acid at position 111 is substituted with tryptophan (W) or an equivalent amino acid.
  • 14. The polypeptide of claim 1, wherein the amino acid at position 36 is substituted with glycine (G) or an equivalent amino acid and the amino acid at position 128 is substituted with tyrosine (Y) or an equivalent amino acid.
  • 15. The polypeptide of claim 1, wherein the amino acid at position 32 is substituted with glutamine (Q) or an equivalent amino acid, and the amino acid at position 40 is substituted with glutamic acid (E) or an equivalent amino acid.
  • 16. A nucleic acid sequence encoding the polypeptide of claim 1.
  • 17. An organism or a host cell comprising the nucleic acid sequence of claim 16.
  • 18. A method for producing pimelic acid, pimelate semialdehyde or 7-aminoheptanoate (7-AHA) or any product of FIG. 2, comprising the step of enzymatically converting pimeloyl-ACP to pimelic acid in the presence of the polypeptide or functional fragment thereof of claim 1.
  • 19. A method for producing pimelic acid, pimelate semialdehyde or 7-AHA or any product of FIG. 2, comprising the steps of culturing the host cell of claim 17 in a suitable medium and recovering the pimelic acid, pimelate semialdehyde or 7-AHA or other product of FIG. 2.
  • 20. A bio-derived pimelic acid, pimelate semialdehyde or 7-AHA or other product of FIG. 2, that is produced by the method of claim 18.
CROSS REFERENCE TO RELATED APPLICATION

The present application claims the benefit of U.S. Provisional Patent Application No. 62/650,629 filed Mar. 30, 2018, which is incorporated herein by reference for all purposes.

Provisional Applications (1)
Number Date Country
62650629 Mar 2018 US