MODIFIED ACTIVIN A

Information

  • Patent Application
  • 20210269495
  • Publication Number
    20210269495
  • Date Filed
    May 14, 2021
    3 years ago
  • Date Published
    September 02, 2021
    2 years ago
Abstract
The purpose of the present invention is to provide a modified activin A.
Description
TECHNICAL FIELD

The present invention relates to activin A comprising a modified amino acid sequence.


BACKGROUND ART

Activin A is a cytokine belonging to the TGFβ (transforming growth factor-β) superfamily. Activin A is a physiologically and industrially useful protein that is involved in development and differentiation as an erythroid differentiation factor and a mesoderm-inducing factor and regulates diverse functions in various cells.


Examples of known methods for producing protein include a production method by transiently expressing a protein of interest in a plant (Non Patent Literature 1: Sainsbury et al., Curr. Opin. Plant Biol., 19, 1-7, 2014) and a method of expressing a target protein in apoplast (Patent Literature 1: JP Laid-Open Publication No. 2005-501558).


However, it has been reported that when a target protein is expressed in a plant, the expressed target protein is degraded by an endogenous plant protease, causing the problem of a reduced yield (Non Patent Literature 2: Mandal MK. et al., Front Plant Sci., 7, 267, 2015). In particular, there are abundant proteases having various spectra (ranges) of specificities in apoplast (Non Patent Literature 3: Pillary P. et al., Bioengineered, 5: 1, 15-20, 2014). Furthermore, the site of the target protein recognized by a protease varies depending on the type of protein, and thus, it is difficult to predict the site. Known methods to address these problems include a method of changing protein localization by using a protein sorting signal and a method of coexpressing a protease inhibitor in a plant (Non Patent Literature 2 and Non Patent Literature 3).


Some conformations of activin A have been reported (Non Patent Literature 4: Wang X. et al., Nat. Commun. 2016 Jul. 4; 7: 12052). The structural information about the site recognized by a protease other than a proprotein convertase (Furin) is unknown.


CITATION LIST
Patent Literature



  • [Patent Literature 1]

  • JP Laid-Open Publication No. 2005-501558



Non Patent Literature



  • [Non Patent Literature 1]

  • Sainsbury et al., Curr. Opin. Plant Biol., 19, 1-7, 2014

  • [Non Patent Literature 2]

  • Mandal MK. et al., Front Plant Sci., 7, 267, 2015

  • [Non Patent Literature 3]

  • Pillary P. et al., Bioengineered, 5: 1, 15-20, 2014

  • [Non Patent Literature 4]

  • Wang X. et al., Nat. Commun. 2016 Jul. 4; 7: 12052

  • [Non Patent Literature 5]

  • Koretz K. et al., Histochemistry, 86: 5, 471-8, 1987



SUMMARY OF INVENTION
Technical Problem

As for activin A, conventionally known methods have not yet solved the problem of degradation by a protease other than a proprotein convertase (Furin) (hereinafter, may be simply referred to as “protease”). Accordingly, a novel activin A that is hard to be degraded by the above described protease (has resistance to degradation by the above described protease) is needed.


Solution to Problem

The present inventors conducted dedicated research to solve the above described problems and have found an unknown recognition site for proteases on activin A. Furthermore, the present inventors modified the amino acid sequence of a proregion of activin A, thereby successfully generating a modified activin A that is hard to be degraded by the protease, and thus, have accomplished the present invention.


Specifically, embodiments of the present invention are as follows:


(1) Activin A having resistance to degradation by a protease other than a proprotein convertase (Furin).


(2) The activin A according to the above described (1), wherein the protease other than a proprotein convertase (Furin) is an endogenous plant protease.


(3) The activin A according to the above described (1) or (2), wherein the activin A comprises a modified proregion.


(4) The activin A according to the above described (3), wherein the amino acid sequence of the modified proregion comprises an amino acid sequence in which at least one amino acid is deleted, substituted and/or added in an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6.


(5) Activin A comprising a modified proregion, wherein the amino acid sequence of the modified proregion comprises an amino acid sequence in which at least one amino acid is deleted, substituted and/or added in the amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6.


(6) The activin A according to the above described (5), wherein the amino acid sequence of the modified proregion comprises any amino acid sequence selected from the following (a) to (d):

    • (a) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is deleted;
    • (b) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a spacer sequence consisting of 1 to 10 amino acids;
    • (c) an amino acid sequence in which at least one of amino acids from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a different amino acid; and
    • (d) an amino acid sequence in which an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 has at least one amino acid inserted therein.


      (7) The activin A according to the above described (6), wherein the amino acid constituting the spacer sequence in the above (b) is at least one amino acid selected from glycine, alanine, and serine.


      (8) The activin A according to the above described (6), wherein the different amino acid in the above (c) is one selected from alanine, serine, glycine, valine, leucine, and isoleucine.


      (9) The activin A according to any of the above described (5) to (8), wherein the activin A has resistance to degradation by a protease other than a proprotein convertase (Furin). Herein, the protease other than a proprotein convertase (Furin) is, for example, an endogenous plant protease.


      (10) The activin A according to the above described (5), wherein the amino acid sequence of the modified proregion comprises any of amino acid sequences selected from the following (e) to (g):
    • (e) an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20;
    • (f) an amino acid sequence in which one or several amino acids are deleted, substituted or added in an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20, and activin A comprising the amino acid sequence has resistance to degradation by a protease other than a proprotein convertase (Furin); and
    • (g) an amino acid sequence having 80% or more homology with an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20, and activin A comprising the amino acid sequence has resistance to degradation by a protease other than a proprotein convertase (Furin).


      (11) A polynucleotide encoding the activin A according to any of the above described (1) to (10).


      (12) A vector comprising the polynucleotide according to the above described (11).


      (13) A transformant expressing the activin A according to any of the above described (1) to (10).


      (14) The transformant according to the above described (13), wherein the transformant is a plant.


      (15) The transformant according to the above described (14), wherein the transformant expresses the activin A in apoplast.


      (16) A method for producing activin A, comprising the step of recovering the activin A expressed in the transformant according to any of the above described (13) to (15).


      (17) A method for producing activin A, comprising the following steps:
    • (a) recovering the activin A expressed in the transformant according to any of the above described (13) to (15); and
    • (b) treating the obtained activin A with a proprotein convertase.


Effects of Invention

The modified activin A of the present invention allows for improving the yield of activin A in producing the activin A in the presence of the protease other than a proprotein convertase (Furin) (for example, in planta), compared to a wild type activin A that is not modified.





BRIEF DESCRIPTION OF DRAWINGS


FIG. 1 is a schematic figure showing the structure of a human activin A.



FIG. 2 is an image demonstrating that degradation of a modified activin A by proteases was suppressed.





DESCRIPTION OF EMBODIMENTS

Hereinbelow, the present invention will be described in detail. The following embodiment is illustrated to describe the present invention and is not intended to limit the present invention to this embodiment. The present invention may be carried out in various modes without departing from the spirits of the present invention. This specification also encompasses the contents described in the specification and drawings of the Japanese patent application (Japanese Patent Application No. 2018-214630) filed on Nov. 15, 2018, to which the present application claims priority.


1. Summary

Activin A is a cytokine belonging to the TGFβ superfamily. Activin A is a physiologically and industrially useful protein that is involved in development and differentiation as an erythroid differentiation factor and a mesoderm-inducing factor and regulates diverse functions in various cells.


An exemplary method for producing a target protein is a method of producing the protein by transiently expressing the same in a plant. However, it has been reported that when a target protein is expressed in a plant, the expressed target protein is degraded by a protease other than a proprotein convertase (Furin) (for example, an endogenous plant protease), causing the problem of a reduced yield. In particular, the abundance of endogenous proteases in apoplast is a major obstacle to expression of the target protein in the apoplast. Furthermore, the site of the target protein recognized by a protease varies depending on the type of protein, and thus, it is difficult to predict the site.


To address these problems, a method of changing protein localization by using a protein sorting signal and a method of coexpressing a protease inhibitor in a plant have been used in the conventional art. However, the problem of degradation by a protease still remains unsolved for activin A.


Under such circumstances, the present inventors have found an unknown recognition site for proteases on activin A; and modified the amino acid sequence of a proregion (also referred to herein as “prodomain”) of activin A, thereby successfully generating a modified activin A that is hard to be degraded by a protease other than a proprotein convertase (Furin) (has resistance to degradation by the protease). Thus, the present inventors have accomplished the present invention. The present invention provides a modified activin A that is hard to be degraded by the protease, by modifying the amino acid sequence of the proregion of activin A. The present inventors have found that activin A becomes hard to be degraded by the protease by modifying an amino acid sequence of a proregion of the activin A without modifying an amino acid sequence of a mature region of the activin A that is physiologically active.


The modified activin A of the present invention allows for improving the yield of activin A in producing the activin A in the presence of the protease other than a proprotein convertase (Furin) (for example, in planta), compared to an unmodified wild type activin A. In this respect, the modified activin A of the present invention is highly useful.


2. Activin A and Modified Form Thereof

Activin A is a cytokine belonging to the TGFβ (transforming growth factor-β) superfamily. Activin A is a protein that is involved in development and differentiation as an erythroid differentiation factor and a mesoderm-inducing factor and regulates diverse functions in various cells.


The present invention provides activin A that is hard to be degraded by a protease other than a proprotein convertase (Furin) (has resistance to degradation by the above protease), by modifying the amino acid sequence of a proregion of activin A. Such activin A allows for improving the yield of activin A in producing the same in the presence of the protease, compared to a wild type activin A that is unmodified.


In the present invention, “activin A” refers to both or either one of pro-activin A and mature activin A. As the term “activin A” is used in the present invention, activin A may be a mixture of pro-activin A and mature activin A or may be either protein. Pro-activin A is a precursor of mature activin A and is a protein comprising a proregion, a recognition region for a proprotein converting enzyme, and a mature region (FIG. 1).


In the present invention, examples of the amino acid sequence of pro-activin A include, but not limited to, an amino acid sequence set forth in SEQ ID NO: 1 (human), SEQ ID NO: 2 (mouse), and SEQ ID NO: 3 (rat). In the present invention, examples of the amino acid sequence of the proregion include, but not limited to, an amino acid sequence set forth in SEQ ID NO: 4 (human), SEQ ID NO: 5 (mouse), and SEQ ID NO: 6 (rat). The amino acid sequence of the recognition region for a proprotein converting enzyme is an amino acid sequence set forth in SEQ ID NO: 7, and the amino acid sequence of the mature region (mature activin A) is an amino acid sequence set forth in SEQ ID NO: 8. Those skilled in the art can readily obtain these amino acid sequences and conformational information from known databases such as GenBank and UniProt Protein Data Bank (PDB). For example, human pro-activin A has GenBank accession number NP_002183.1 and PDB accession number 5HLY.


Mature activin A is a protein comprising a mature region, the protein being generated when pro-activin A is cleaved by a proprotein converting enzyme (Furin). Mature activin A is a homodimer formed by linking 116-amino acid residue βA chains by a disulfide bond.


In the present invention, the type of animals from which activin A is derived is not limited. Examples of the animals include a human, a mouse, a rat, a guinea pig, a rabbit, a cat, a dog, a pig, a monkey, and a cow. Preferably, the animal is a human. The amino acid sequences of the mature region of activin A have 100% homology among humans, mice, rats, cats, pigs, and cows.


The present invention provides activin A that has resistance to degradation by a protease other than a proprotein convertase (Furin). In the present invention, a “protease other than a proprotein convertase (Furin)” is not limited, as long as the protease has the activity of degrading (for example, cleaving) activin A. The protease in the present invention may be an endogenous protease comprised in a transformant of a host such as a plant, a non-human mammal and a cell derived therefrom, a bacterium, a yeast, and a fungus, or may be an exogenous protease, which is found outside the transformant. The protease is preferably an endogenous plant protease or an exogenous plant protease, and more preferably an endogenous plant protease. Specifically, there are abundant proteases having various spectra (ranges) of specificities in a plant body (for example, apoplast) (Pillary P. et al., Bioengineered, 5: 1, 15-20, 2014), and thus, the endogenous plant protease is not limited, as long as the protease has the activity of degrading activin A.


Those skilled in the art can determine whether a specific protease has the activity of degrading activin A by using a known method such as SDS-PAGE and Western blotting.


In the present invention, the phrase “have resistance to degradation by a protease other than a proprotein convertase (Furin)” means that, in particular, when pro-activin A was expressed in a transformant, extracted, and purified as necessary and then the resulting protein was analyzed, the percentage of pro-activin A content relative to the total resulting protein is usually 70% or more, preferably 80% or more, and more preferably 84% or more. This phrase does not necessarily mean that the percentage of the pro-activin A content has to be 100% (not necessarily mean that degradation has to be suppressed by 100%). The amount or percentage of pro-activin A content can be measured by Western blotting as described in Examples. Thus, those skilled in the art can determine whether the modified activin A has resistance to degradation by a protease other than a proprotein convertase (Furin) by measuring the amount or percentage of the pro-activin A content relative to the total protein by Western blotting.


In the present invention, “protease other than a proprotein convertase (Furin)” may be expressed as “protease (excluding a proprotein convertase (Furin).”


One embodiment of the present invention is activin A comprising a modified proregion. A modified proregion is a proregion in which a sequence expected as a loop region is modified, and is preferably a proregion in which, among loop regions in the proregion, at least one sequence expected as a loop region has been deleted. Herein, the loop region is a part having a loop structure, and more specifically, a part from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6.


In the present invention, the amino acid sequence of the modified proregion comprises an amino acid sequence in which at least one amino acid is deleted, substituted and/or added in an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 (activin A comprising a modified proregion is also referred to herein as “modified activin A”).


The present invention also provides activin A comprising a modified proregion, wherein the amino acid sequence of the modified proregion comprises an amino acid sequence in which at least one amino acid is deleted, substituted and/or added in an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6, and the activin A has resistance to degradation by a protease other than a proprotein convertase (Furin).


Examples of the amino acid sequence of the modified proregion include, but not limited to, the following amino acid sequences (a) to (d):

    • (a) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is deleted;
    • (b) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a spacer sequence consisting of 1 to 10 amino acids;
    • (c) an amino acid sequence in which at least one of amino acids from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a different amino acid; and
    • (d) an amino acid sequence in which an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 has at least one amino acid inserted therein.


Furthermore, examples of the amino acid sequence of the modified proregion include, but not limited to, amino acid sequences which are the following amino acid sequences (a) to (d) and wherein activin A comprising the amino acid sequence has resistance to degradation by a protease other than a proprotein convertase (Furin),

    • (a) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is deleted;
    • (b) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a spacer sequence consisting of 1 to 10 amino acids;
    • (c) an amino acid sequence in which at least one of amino acids from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a different amino acid; and
    • (d) an amino acid sequence in which an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 has at least one amino acid inserted therein.


The above described amino acid sequence (a) is the amino acid sequence set forth in any of SEQ ID NO: 9 (human), SEQ ID NO: 10 (mouse), and SEQ ID NO: 11 (rat).


In the amino acid sequence (b), a “spacer sequence” is an amino acid sequence consisting of 1 to 10 amino acids. The amino acid constituting the spacer sequence is at least one selected from glycine, alanine, and serine. Specifically, the spacer sequence may be an amino acid sequence composed of only one type of amino acid, such as only glycine or only alanine; or may be an amino acid sequence composed of two or more types of amino acids, for example: glycine and alanine; glycine and alanine and leucine. The number of amino acids in the spacer sequence is 1 to 10, preferably 1 to 5, more preferably 1 to 4, and even more preferably 1 to 3. More specifically, examples of the spacer sequence include -G-, -GG-, -GGG-, and -GGGG-. In the present invention, when a spacer consists of one amino acid residue, “spacer sequence” may also be referred to as “spacer residue.”


Those skilled in the art can search for the type and number of amino acid candidates as well as amino acid sequence candidates that constitute the spacer sequence by using molecular visualization software described below. Furthermore, in the present invention, a substructure can be inserted instead of the spacer sequence, provided that the substructure is registered in PDB, has ends whose relative coordinates correspond to the coordinates of an insertion site on activin A, can form a peptide bond, and does not cause a steric hindrance after insertion.


Examples of the above described amino acid sequence (b) include, but not limited to, an amino acid sequence set forth in any of SEQ ID NO: 12 (human), SEQ ID NO: 13 (mouse), and SEQ ID NO: 14 (rat) that comprises one glycine as the spacer sequence; an amino acid sequence set forth in any of SEQ ID NO: 15 (human), SEQ ID NO: 16 (mouse), and SEQ ID NO: 17 (rat) that comprises two glycines as the spacer sequence; and an amino acid sequence set forth in any of SEQ ID NO: 18 (human), SEQ ID NO: 19 (mouse), and SEQ ID NO: 20 (rat) that comprises three glycines as the spacer sequence.


In the above amino acid sequence (c), a “different amino acid” is not limited as long as the amino acid does not cause a steric hindrance in activin A. Examples thereof include alanine, serine, glycine, valine, leucine, and isoleucine.


In the above amino acid sequence (d), the amino acid to be inserted is not limited as long as the amino acid does not cause a steric hindrance in activin A. Examples thereof include alanine, serine, glycine, valine, leucine, and isoleucine.


In the present invention, in addition to the above described amino acid sequence set forth in any of SEQ ID NOs: 9 to 20, the following amino acid sequences can be used as an amino acid sequence of the modified proregion:

    • (f) an amino acid sequence in which one or several amino acids are deleted, substituted or added in an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20, and activin A comprising the amino acid sequence has resistance to degradation by a protease other than a proprotein convertase (Furin); and
    • (g) an amino acid sequence having 80% or more homology with an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20, and activin A comprising the amino acid sequence has resistance to degradation by a protease other than a proprotein convertase (Furin).


In the above amino acid sequence (f), examples of an amino acid sequence in which one or several amino acids are deleted, substituted or added in set forth in any of SEQ ID NOs: 9 to 20 include the following amino acid sequences:

    • (i) an amino acid sequence in which 1 to 10 (for example, 1 to 5, preferably 1 to 3, more preferably 1 to 2, and even more preferably 1) amino acids of an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20 is deleted;
    • (ii) an amino acid sequence in which 1 to 10 (for example, 1 to 5, preferably 1 to 3, more preferably 1 to 2, and even more preferably 1) amino acids of an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20 is substituted with a different amino acid;
    • (iii) an amino acid sequence in which 1 to 10 (for example, 1 to 5, preferably 1 to 3, more preferably 1 to 2, and even more preferably 1) amino acids are added to an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20; and
    • (iv) an amino acid sequence mutated by a combination of the above described (i) to (iii).


In the above amino acid sequence (g), examples of the amino acid sequence having 80% or more homology with an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20 include an amino acid sequence having 80% or more, 90% or more, 95% or more, 98% or more, or 99% or more homology with an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20.


Those skilled in the art can readily examine homology among amino acid sequences by using a known database such as FASTA or BLAST.


In the present invention, “resistance to degradation by a protease other than a proprotein convertase (Furin)” means the property that makes activin A hard to be degraded by a protease other than a proprotein convertase (Furin). Furthermore, as described above, the phrase “have resistance to degradation by a protease other than a proprotein convertase (Furin)” in the present invention means that, in particular, when pro-activin A was expressed in a transformant, extracted, and purified as necessary and then the resulting protein was analyzed, the percentage of pro-activin A content relative to the total resulting protein is usually 70% or more, preferably 80% or more, and more preferably 84% or more. This phrase does not necessarily mean that the percentage of the pro-activin A content has to be 100% (not necessarily mean that degradation has to be suppressed by 100%). Those skill in the art can determine whether a modified activin A has resistance to degradation by a protease other than a proprotein convertase (Furin) by measuring the amount or percentage of the pro-activin A content relative to the total protein by Western blotting, as described in Examples.


The present invention also provides activin A comprising a modified proregion, wherein the amino acid sequence of the modified proregion comprises an amino acid sequence in which at least one amino acid has been deleted, substituted and/or added in an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6, and wherein the activin A has the activity of inducing differentiation into an erythroblast when cleaved by a proprotein converting enzyme.


Examples of the amino acid sequence of the modified proregion include, but not limited to, the below described amino acid sequences (a) to (g). Examples of the activin A comprising a modified proregion include, but not limited to, the activin A comprising any of the below described amino acid sequences (a) to (g) and having the activity of inducing differentiation into an erythroblast when cleaved by a proprotein converting enzyme.

    • (a) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is deleted;
    • (b) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a spacer sequence consisting of 1 to 10 amino acids;
    • (c) an amino acid sequence in which at least one of amino acids from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a different amino acid;
    • (d) an amino acid sequence in which an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 has at least one amino acid inserted therein;
    • (e) an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20;
    • (f) an amino acid sequence in which one or several amino acids are deleted, substituted or added in an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20; and
    • (g) an amino acid sequence having 80% or more homology with an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20.


In the present invention, “has the activity of inducing differentiation into an erythroblast when cleaved by a proprotein converting enzyme” means that activin A resulting from cleavage by a proprotein converting enzyme (a mixture of pro-activin A and mature activin A or mature activin A) has the activity of inducing differentiation of any erythroblastic cultured cell (for example, a F5-5 cell and a K562 cell) into an erythroblast.


The activity of inducing differentiation into an erythroblast can be evaluated by a known method. An exemplary method comprises adding a test polypeptide (test activin A) to the culture solution of erythroblastic cultured cells, staining the cells after cultivation with aminoethylcarbazole (AEC) or the like, and measuring the ED50 value of the test activin A to determine the differentiation rate.


In the present invention, examples of the amino acid sequence of activin A comprising a modified proregion include, but not limited to, amino acid sequences of activin A comprising each of the amino acid sequences set forth in SEQ ID NOs: 9 to 20 (amino acid sequences set forth in SEQ ID NOs: 21 to 32).


The conformation of activin A with modification (deletion, substitution, addition, or a combination thereof) can be analyzed and simulated by using molecular visualization software that displays the structure of a protein in a 3D image based on data in PDB or the like. Examples of such software include Chimera and PyMOL. For example, the above amino acid sequence (a), in which the amino acid sequence from position 182 to 199 has been deleted and no spacer sequence has been inserted, can be subjected to simulation based on an in silico analysis using Chimera to find out whether a steric hindrance occurs. For the above amino acid sequence (b), the above software can be used to search for the type and number of amino acids or amino acid sequence candidates that constitute the spacer sequence. Similarly, the above amino acid sequences (c) and (d) can be subjected to simulation to find out whether a steric hindrance or the like occurs when substitution or insertion by a different amino acid was introduced in the amino acid sequence from position 182 to 199. Thus, those skilled in the art can select appropriately an amino acid residue that causes no steric hindrance when being deleted, substituted and/or added in activin A by using such molecular visualization software.


Furthermore, the chemical property or physical property of amino acids (polar/non-polar, acidic/basic, hydrophilic/hydrophobic, side chain size, and the like) is known. Those skilled in the art can appropriately select an amino acid to be deleted, substituted, and/or added on the basis of these properties. Substitution between amino acids that have similar structures or properties is predicted to be hard to substantially inhibit the biological activity of a polypeptide.

    • 1) Polar amino acids: lysine, arginine, histidine, aspartic acid, glutamic acid, serine, threonine, asparagine, glutamine, tyrosine, and cysteine;
    • 2) Nonpolar amino acids: glycine, alanine, valine, leucine, isoleucine, phenylalanine, tryptophan, methionine, and proline;
    • 3) Acidic amino acids: aspartic acid and glutamic acid;
    • 4) Basic amino acids: lysine, arginine, and histidine;
    • 5) Neutral amino acids: glycine, alanine, valine, leucine, isoleucine, phenylalanine, tyrosine, tryptophan, serine, threonine, asparagine, glutamine, methionine, cysteine, and proline;
    • 6) Aliphatic amino acids: glycine, alanine, valine, leucine, and isoleucine;
    • 7) Aromatic amino acids: phenylalanine, tyrosine, and tryptophan;
    • 8) Amino acids having a hydroxyl group: serine and threonine;
    • 9) Amino acids having an amido group: asparagine and glutamine;
    • 10) Sulfur-containing amino acids: methionine and cysteine;
    • 11) Hydrophobic amino acids: glycine, alanine, valine, leucine, isoleucine, phenylalanine, tyrosine, tryptophan, methionine, cysteine, and proline (it is noted that glycine, tyrosine, tryptophan, and cysteine may be classified as hydrophilic amino acids);
    • 12) Hydrophilic amino acids: lysine, arginine, histidine, aspartic acid, glutamic acid, asparagine, glutamine, serine, and threonine.


3. Polynucleotide, Vector, and Transformant
(1) Polynucleotide

The nucleotide sequence of a polynucleotide of the present invention is not limited as long as the sequence is DNA or RNA encoding a modified activin A of the present invention. Those skilled in the art can optimize codons for the polynucleotide depending on the type of host in which activin A is expressed. This optimization can improve the amount of the modified activin A expressed in the host. Examples of the polynucleotide encoding the modified activin A of the present invention include, but not limited to, a polynucleotide comprising or consisting of a nucleotide sequence set forth in any of SEQ ID NOs: 33 to 36 (all thereof are human nucleotide sequences).


In addition to the polynucleotide comprising or consisting of a nucleotide sequence set forth in any of SEQ ID NOs: 33 to 36, a polynucleotide described below can be used as the polynucleotide of the present invention. Such a polynucleotide is a polynucleotide that hybridizes with a polynucleotide consisting of a nucleotide sequence complementary to a nucleotide sequence set forth in any of SEQ ID NOs: 33 to 36 under stringent conditions and encodes activin A having resistance to degradation by a protease other than a proprotein convertase (Furin). In the present invention, a “stringent condition” may be either a low stringency condition, a moderate stringency condition, or a high stringency condition. A “low stringency condition” is, for example, the following condition: 5×SSC, 5×Denhardt's solution, 0.5% SDS, 50% formamide, and 32° C. A “medium stringency condition” is, for example, the following condition: 5×SSC, 5×Denhardt's solution, 0.5% SDS, 50% formamide, and 42° C. A “high stringency condition” is, for example, the following condition: 5×SSC, 5×Denhardt's solution, 0.5% SDS, 50% formamide, and 50° C. For a detailed procedure of a hybridization method, one can refer to “Molecular Cloning, A Laboratory Manual (4th edition)” (Cold Spring Harbor Laboratory Press (2012)), and the like.


As the polynucleotide of the present invention, a polynucleotide that has 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, 95% or more, 98% or more, or 99% or more homology with the nucleotide sequence set forth in any of SEQ ID NOs: 33 to 36, and encodes activin A having resistance to degradation by a protease other than a proprotein convertase (Furin) can also be used.


Methods for introducing a mutation into a polynucleotide are known. For example, those skilled in the art can introduce a mutation into the polynucleotide of the present invention by using a known method including a site-directed mutagenesis strategy such as the Kunkel method and the Gapped duplex method, the overlap extension PCR method and the QuikChange method. The amino acid sequence of activin A can be modified by introducing a mutation into the polynucleotide of the present invention.


In the present invention, an affinity tag can be added to activin A for affinity-purifying activin A. Examples of usable affinity tags include, but not limited to, a histidine tag (an amino acid sequence consisting of contiguous histidine residues), a GST tag, a FLAG tag, and a c-myc tag. Methods for adding a histidine tag are known and those skilled in the art can readily add a histidine tag to the N-terminus of the activin A of the present invention by using a known method. One example of the amino acid sequences of activin A with a histidine tag added to the N-terminus thereof is set forth in SEQ ID NO: 37.


In the present invention, a signal sequence can also be added to activin A. Examples of the signal sequence include a KDEL signal. Methods for adding a signal sequence are known. A sequence optimized for expression thereof in a plant cell can be used.


(2) Vector

In the present invention, a “vector” is not limited as long as the vector comprises the polynucleotide of the present invention described in the above (1). For example, a plasmid vector, a viral vector, an Agrobacterium bacterium, or the like can be used. When a plant is used as a host, for example, a plant viral vector, an Agrobacterium bacterium, or the like can be used as the vector. More specifically, for example, a TMV-based vector, a PVX-based vector, a CPMV-based vector, a CMV-based vector, a PPV-based vector, an AIMV-based vector, a ZYMV-based vector, or the like can be used. The vector used in the present invention may be either a vector for transient expression or a vector for constitutive expression. Those skilled in the art can appropriately select a vector to be used depending on the type of host into which the vector is introduced and the purpose.


In the vector of the present invention, an additional sequence may be linked to the polynucleotide of the present invention, as desired. Examples of the additional sequence include a cis element such as an enhancer, a splicing signal, a poly(A) addition signal, a ribosome binding sequence (SD sequence), a selective marker gene, and a reporter gene. Examples of the selective marker gene include a dihydrofolate reductase gene, an ampicillin-resistant gene, and a neomycin-resistant gene. Examples of the reporter gene include genes for green fluorescent protein (GFP) or variants thereof (fluorescent proteins such as EGFP, BFP, and YFP), luciferase, alkaline phosphatase, and LacZ.


(3) Transformant

In the present invention, a transformant can be obtained by introducing the vector described in the above (2) into a host. In the present invention, a “transformant” is a non-human transformant. In the present invention, a “host” is an organism into which the vector of the present invention is introduced and in which a modified activin A of interest is expressed. The host is not limited as long as the host expresses the modified activin A of the present invention. For example, a plant, a non-human mammal and a cell derived therefrom, a bacterium, a yeast, a fungus, or the like can be used, and a plant is preferred. Examples of the plant include a Nicotiana plant, a moss plant (for example, Physcomitrella patens (P. patens)), a potato (for example, S. tuberosum), a gramineous plant (for example, O. sativa), a cruciferous plant, and lettuce; and a Nicotiana plant is preferred. Examples of the Nicotiana plant include, but not limited to, Nicotiana benthamiana (N. benthamiana), Nicotiana tabacum (N. tabacum), and Nicotiana excelsior (N. excelsior). Examples of the non-human mammal include, but not limited to, a mouse, a rat, a guinea pig, a rabbit, a cat, a dog, a pig, a monkey, and a cow, and a cell derived therefrom. Examples of the bacterium include, but not limited to, E. coli.


Introduction of a vector into a host can be performed by using a known method. Examples of the known gene introduction method include a method using a viral vector, an agroinfection method, a calcium phosphate method, a microinjection method, a particle gun method, a DEAE-dextran method, electroporation, and a cationic lipid-mediated method. When the host is a plant, the method using a viral plant vector and the agroinfection method are preferred. The vectors that can be used in these methods are as described in the above (2) as “a vector when a plant is used as a host.”


In the present invention, when a plant is used as a host, the plant is cultivated before transformation. A method for cultivating plants is as described below.


First, seeds are sown on a seedling tray supplemented with a fertilizer, and the plant after sowing is grown for several days in an artificial climate chamber with a controlled light cycle. When a liquid fertilizer is used as the fertilizer, a urethane mat for hydroponics can be impregnated with the liquid fertilizer, and then placed in the seedling tray.


Then, the grown plant body is transplanted to a panel for cultivation (first period), the panel after transplantation is placed in the artificial climate chamber, and cultivation is performed for several days by, for example, a deep flow technique (DFT system).


Subsequently, the plant body is taken out from the panel for cultivation (first period) and planted on a panel for cultivation (second period). The panel for cultivation (second period) after transplantation is placed in the artificial climate chamber and cultivation is performed for several days by the DFT system to obtain the plant body.


In the above described method, a liquid fertilizer can be used as the fertilizer, but the fertilizer is not limited thereto. When a liquid fertilizer is used, the urethane mat for hydroponics can be impregnated with the liquid fertilizer, and then placed in the seedling tray.


In the present invention, liquid fertilizers are not limited, and commercially available liquid fertilizers can be used in appropriate combinations. The liquid fertilizer can be dissolved in dechlorinated water. Furthermore, the electric conductivity and pH of the liquid fertilizer can be adjusted before use. Those skilled in the art can adjust these by a known method.


In the present invention, examples of the environmental condition include, but not limited to, a condition with a temperature of 10 to 40° C. (for example, 28° C.), a relative humidity of 60 to 80%, a CO2 concentration of 300 to 5000 ppm (for example, 400 ppm or 500 ppm), and the number of days of cultivation for the first period of 0 to 35 days (for example, 9 days) and that for the second period of 0 to 35 days (for example, 7 days). Those skilled in the art can adjust these parameters as appropriate depending on, for example, the growth status of the plant.


In the present invention, as hydroponic techniques, the deep flow technique (DFT system) and a nutrient film technique (NFT system) can be mainly used.


As described above, when the host is a plant, the method using a plant viral vector and the agroinfection method can be used. These methods are well known to those skilled in the art. By way of example, a brief description of the agroinfection method is as follows.


First, the vector of the present invention described in the above (2) is introduced into an Agrobacterium bacterium by electroporation, thereby transforming the Agrobacterium bacterium. Examples of Agrobacterium bacterium that can be used in the present invention include, but not limited to, the GV3101 strain, the LBA4404 strain, the EHA101 strain, the EHA105 strain, and the AGL1 strain.


Then, plant leaves or the like are infected with the transformed Agrobacterium bacterium. Examples of methods for infecting a plant with an Agrobacterium bacterium include a vacuum infiltration method, a syringe infiltration method, a leaf disc method, and a foliar application method. An exemplary procedure for using the vacuum infiltration method is as follows. First, the cultivated plant is turned upside down and immersed in the bacterial suspension of Agrobacterium bacteria in a beaker such that all the leaves are completely immersed in the suspension. Subsequently, the beaker is placed in a vacuum desiccator and allowed to stand for several minutes (for example, one minute) under reduced pressure. Then, the valve is fully opened to restore pressure. After completing pressure restoration, the plant is returned to an upright position and planted in the artificial climate chamber. After infection, the plant is cultivated for 1 to 14 days (for example, 6 days) in the artificial climate chamber by, for example, the DFT system. The environmental condition is similar to that described above, and those skilled in the art can adjust these parameters as appropriate depending on, for example, the growth status of the plant.


In this way, a transformant of the plant can be generated. In infecting a plant with the Agrobacterium bacterium, the plant may be coinfected with multiple types of Agrobacterium bacteria, each comprising a different vector. In this case, Agrobacterium bacteria comprising a vector other than the vector of the present invention may be used in combination. Examples of such vector include a vector comprising a signal peptide of a barley α-amylase or a rice α-amylase, and a PhiC31 integrase expression vector.


Transformants from other hosts can also be generated readily by those skilled in the art by a known method described in, for example, “Molecular Cloning, A Laboratory Manual (4th edition)” (Cold Spring Harbor Laboratory Press (2012)).


4. Method for Producing Activin A

In the present invention, activin A can be produced by recovering activin A from the transformant of the present invention described in the above section 3. Furthermore, in the method of the present invention, a mature region of activin A in which a proregion is removed (mature activin A) can be obtained by purifying the recovered activin A (pro-activin A) and treating the purified pro-activin A with a proprotein convertase (for example, Furin).


The type of transformant used in the method for producing activin A is not limited. By way of example, when the transformant is a plant, the method for producing activin A is as described below.


First, leaves are collected from the transformed plant body that was cultivated for 1 to 14 days (for example, 6 days) after infection with Agrobacterium bacterium, and activin A (pro-activin A) is extracted by using an extraction buffer. The quantity of leaves of the plant body to be collected depends on the type of plant body. The leaves can be frozen and stored at −80° C. before extraction. Examples of the extraction buffer include, but not limited to, phosphate buffer, Tris buffer, and acetate buffer. The pH is usually adjusted to a range between pH 2 and 11, including the range in which the above described buffers work properly.


Next, activin A comprised in the extract is purified. Purification can be performed by using conventional methods alone or in appropriate combinations. The conventional methods are, for example, aqueous two-phase partition, ammonium sulfate fractionation, affinity chromatography, ion exchange chromatography, gel filtration chromatography, hydrophobic chromatography, or reverse-phase chromatography.


Conventional methods can be used to confirm that the resulting purified substance is the target protein, activin A. Examples of the conventional methods include SDS-polyacrylamide gel electrophoresis, N-terminal amino acid sequence analysis, Western blotting, enzyme immunoassay (ELISA), and mass spectrometry.


In this way, purified activin A (pro-activin A) can be obtained.


Furthermore, the mature region of activin A in which a proregion is removed (mature activin A) can be obtained by treating the purified pro-activin A with a proprotein convertase (Furin). A mixture of the proregion and the mature activin A may also be used for experiments.


The activity of the obtained mature activin A (or the mixture of the proregion and the mature activin A) can be evaluated, for example by cultivating a F5-5 cell line (Friend erythroblastic leukemia cell line) in a culture medium supplemented with the mature activin A, then staining the cells with AEC (aminoethylcarbazole), and measuring the ED50 value (indicating the concentration of mature activin A at which the differentiation rate is 50%) of the mature activin A for the differentiation rate.


5. Composition Comprising Activin A

The present invention can provide a composition comprising a modified activin A of the present invention. The composition of the present invention may comprise known additives such as saline, buffer, and an excipient, in addition to the modified activin A.


Hereinbelow, the present invention will be described in detail by way of examples, but the present invention is not limited to these examples.


Example 1
1. Designing Modified Activin A

In this example, a sequence from which a specific sequence of a proregion was removed was designed.


When pro-activin A, which was transiently expressed by a plant, was extracted and then subjected to Western blotting, a plurality of unexpected degradation products was found. Thus, a plurality of sequences to induce site-specific mutations was designed and mutations were induced by using QuikChange (Agilent Technologies, Inc.). The mutated protein was overexpressed and examined in the same way as the above described original sequence. As a result, it was confirmed that the degradation was reduced in more than one sequences. The sequences were a sequence from which Pro182-Gly192 was removed. The Pro182-Gly192 was predicted to be a loop region of the prodomain. PDB 5HLY was used to adjust the dihedral angle between H181 and E200 and these residues were linked by a peptide bond. As a result, it was found not to cause a steric hindrance, and thus, this protein was designed as Mut3-1. Furthermore, an appropriate linker sequence was introduced between the above H181 and E200. A sequence into which (Gly)n (n=1 to 4) was introduced was designed to perform expression screening. Based on SuperLooper (http://bioinf-applied.charite.de/superlooper/description.php), the above linkers (Gly)n (n=2 or 3) that have the relative position coordinates of the residues, H181 and E200 for linker introduction were extracted from the registered PDB structure information (3DZM, 2VK3, and 5AA5 having GG, and 4GNO having GGG). Thus, sequences of Mut3-2 (n=1), Mut3-3 (n=2), and Mut3-4 (n=3) were designed.


Example 2
Production of Modified Activin A

On the basis of the information of the modified activin A designed in Example 1, the modified activin A was produced by a below described method.


1. Preparation of Vector

A vector for expressing pro-activin A in which a histidine tag (HHHHHH) was added to the N-terminus thereof was generated as described below. A gene encoding pro-activin A in which a histidine tag was added to the N-terminus thereof was inserted into a Bsa I site of a tobacco mosaic virus (TMV) 3′-provector (pICH31070, NOMAD Bioscience GmbH) by Golden Gate cloning method (Engler et al., 2008), thereby generating a pN-His-Pro-activin.


Next, a TMV 3′-provector that expresses a gene encoding a modified pro-activin A was generated as described below. PCR was carried out by using QuikChange Lightning Kit (Agilent Technologies, Inc.) with the pN-His-Pro-activin as a template and the following primers for mutation introduction, thereby generating a pMut3-1.









(SEQ ID NO: 38)


5′-CCAACAGCAGAAGCACGAGAGATCTGAGTTGC-3′





(SEQ ID NO: 39)


5′-GCAACTCAGATCTCTCGTGCTTCTGCTGTTGG-3






2. Generation of Transformant
(1) Cultivation of Host Plant

In this example, Nicotiana benthamiana, which is a Nicotiana plant, was used as a host plant.


(1-1) Sowing

A urethane mat for hydroponics (Ematsu Kasei; (W)587.5 mm×(D)282 mm×(H)28 mm; 12×2 blocks; hole diameter (ϕ): 9 mm) was impregnated with a liquid fertilizer for sowing (0.78 g/L Otsuka House No. S1 (Otsuka AgriTechno Co., Ltd.) and 0.25 g/L Otsuka House No. 2 (Otsuka AgriTechno Co., Ltd.); pH 5.0) and placed in a seedling tray ((W)600 mm×(D)300 mm×(H)300 mm). Then, seeds of Nicotiana benthamiana were sown on the mat.


(1-2) Raising Seedlings

The plant after sowing was grown in an artificial climate chamber (NC-410HC) (Nippon Medical & Chemical Instruments Co., Ltd.) at a room temperature of 28° C. with a light cycle of 16 hours of light and 8 hours of darkness for 12 days.


(1-3) Cultivation (First Period)

The urethane mat used for raising seedlings was divided into each block and transplanted to a panel for cultivation (first period) ((W)600 mm×(D)300 mm, 30 holes). The panel for cultivation (first period) after transplantation was placed in the artificial climate chamber (LH-410SP) (Nippon Medical & Chemical Instruments Co., Ltd.) and cultivation was performed for 9 days by a deep flow technique (DFT system). The environmental conditions and the liquid fertilizer conditions were controlled as follows.


«Environmental Conditions»
Temperature: 28° C.

Relative humidity: 40 to 60%


CO2 concentration: 400 ppm


Lighting: average photosynthetic photon flux density (PPFD): 140 μmol·m−2·sec−1, 24 hours continuous lighting, a three band fluorescent lamp “Rupika Line” (Mitsubishi Electric Corporation)


«Liquid Fertilizer Conditions»

Fertilizer liquid A (150 g/L Otsuka House No. S1 and 2.5 g/L Otsuka House No. 5 (Otsuka AgriTechno Co., Ltd.)) and fertilizer liquid B (100 g/L Otsuka House No. 2) were dissolved individually in dechlorinated water and these solutions were mixed in equal proportions. The resulting mixture was used as the liquid fertilizer. A pH adjuster “Down” (Otsuka AgriTechno Co., Ltd.) and 4% aqueous KOH solution were used to adjust pH. The electrical conductivity (EC) and the pH of the liquid fertilizer were adjusted to an EC of 2.3 mS/cm and pH 6.0, respectively, by using “Easy Fertilizer Controller RAKU-RAKU 3” (CEM Corporation).


(1-4) Cultivation (Second Period)

The plant body was taken out from the panel for cultivation (first period) and planted on a panel for cultivation (second period) ((W)600 mm×(D)300 mm, 6 holes). The panel for cultivation (second period) after transplantation was placed in the artificial climate chamber (LH-410SP) (Nippon Medical & Chemical Instruments Co., Ltd.) and cultivation was performed for 7 days by the DFT system (for 28 days after sowing). Environmental conditions were controlled as described below.


«Environmental Conditions»
Temperature: 28° C.

Relative humidity: 60 to 80%


CO2 concentration: 500 ppm


Lighting: average photosynthetic photon flux density (PPFD): 140 mol·m−2·sec−1, 24 hours continuous lighting, a three band fluorescent lamp “Rupika Line” (Mitsubishi Electric Corporation)


(2) Generation of Transformant
(2-1) Infection by Vacuum Infiltration

The vectors of the present invention generated in the above “1.” were individually introduced into an Agrobacterium GV3101 strain by electroporation. Then, agroinfiltration was used to coinfect Nicotiana benthamiana with each of the obtained GV3101 strains: a GV3101 strain carrying a 5′-provector comprising a signal peptide of rice α-amylase (pICH20155, NOMAD Bioscience GmbH) and a GV3101 strain carrying a PhiC31 integrase expression vector (pICH14011, NOMAD Bioscience GmbH).


Specifically, the Nicotiana benthamiana obtained on Day 28 after sowing in the above section “2.(1)” was turned upside down and immersed in the bacterial suspension of Agrobacterium bacteria in a beaker such that all the leaves were completely submerged in the suspension.


Subsequently, the beaker was placed in a vacuum desiccator (FV-3P) (Tokyo Glass Kikai Co., Ltd.) and allowed to stand for one minute under reduced pressure at −0.09 MPa. Then, the valve was fully opened to restore pressure.


After completing pressure restoration, the plant was returned to an upright position and planted in the artificial climate chamber (LH-410SP) (Nippon Medical & Chemical Instruments Co., Ltd.).


(2-2) Cultivation of Infected Leaves (Expression Step)

Cultivation after infection was performed by using the artificial climate chamber (LH-410SP) (Nippon Medical & Chemical Instruments Co., Ltd.). Cultivation was performed for six days by the DFT system. Environmental conditions were controlled as described below.


«Environmental Conditions»
Temperature: 28° C.

Relative humidity: 60 to 80%


CO2 concentration: 500 ppm


Lighting: average photosynthetic photon flux density (PPFD): 140 μmol·m−2·sec−1, 24 hours continuous lighting, a three band fluorescent lamp “Rupika Line” (Mitsubishi Electric Corporation)


3. Production of Modified Pro-Activin A
(1) Extraction

Tobacco leaves were harvested from the transformed Nicotiana benthamiana six days after infection. The leaves were frozen and stored at −80° C. until extraction.


A buffer comprising 0.1 M sodium phosphate, 0.5 M arginine, and 5 mM sodium sulfite, pH 8.0 (or a phosphate buffer) was used as an extraction buffer.


(2) Purification
(2-1) Purification by Ammonium Sulfate Fraction

Ammonium sulfate was added to the supernatant collected by the above described method to produce a 35% saturated ammonium sulfate solution. After stirring the mixture at room temperature for one hour, the mixture was centrifuged at 15,000×g for 15 minutes at room temperature to collect the supernatant of 35% ammonium sulfate fractionation. Subsequently, ammonium sulfate was added thereto to produce a 60% saturated ammonium sulfate solution and stirred at room temperature for one hour. This solution was centrifuged at 15,000×g for 15 minutes to collect precipitates of the 60 to 90% ammonium sulfate fractionation.


(2-2) Purification by Affinity Chromatography

To 50 g of precipitates of the 60 to 90% ammonium sulfate fractionation from activin A-expressing tobacco leaves, 10 mL of equilibration solution for histidine (His)-tag affinity purification (20 mM HEPES, 150 mM NaCl, 10% (w/v) glycerol, pH 8.0) was added, and the precipitate was dissolved. This solution was passed through a HisTrap HP 1 mL column (GE Healthcare) equilibrated with the equilibration solution for His-tag affinity purification at a speed to achieve retention time of 3 minutes. Then, the column was washed with a washing solution for His-tag affinity purification (20 mM HEPES, 150 mM NaCl, 20 mM imidazole, 10% (w/v) glycerol, pH 8.0). Lastly, an elution solution for His-tag affinity purification (20 mM HEPES, 150 mM NaCl, 200 mM imidazole, 10% (w/v) glycerol, pH 8.0) was passed through the column and the elution peak was collected to obtain a modified pro-activin A.


4. Production of Mature Activin A

A proprotein convertase (Furin) was added to the modified pro-activin A obtained by the above described method (Mut3-1, Mut3-2, Mut3-3, and Mut3-4 (comprising the amino acid sequences of SEQ ID NOs: 9, 12, 15, and 18, respectively)) under conditions of 25 mM Tris-HCl, pH 8.0, 10% glycerol, and 1 mM CaCl2 to obtain a mature activin A.


In this example, activin A in which an amino acid sequence of a proregion was modified (a modified pro-activin A and a mature activin A) was successfully produced.


Example 3
1. Functional Analysis of Modified Pro-Activin A

The purified modified pro-activin A obtained in “3. Production of Modified Pro-activin A” of Example 2 was analyzed by Western blotting to determine how a plant endogenous protease degraded the protein. Specifically, Penta-His Antibody HRP Conjugate (QIAGEN) was used as an antibody and the samples comprising each modified pro-activin A were subjected to Western blotting. LumiGLO Reagent and Peroxide (Cell Signaling Technology, Inc.) was used as a luminescent reagent and was detected by ImageQuant LAS 500 (GE Healthcare). The obtained Western blotting image is shown in FIG. 2. Wild-type Activin A (N-His-Activin A in the figure) (SEQ ID NO: 37) was used as a control. Densitometric analysis was also performed by using analysis software ImageJ and the results are shown in Table 1. In Table 1, “other polypeptides” are peptides resulting from degradation of the expressed proteins by the plant endogenous protease.














TABLE 1






N-His-






Expressed protein
Activin A
Mut3-1
Mut3-2
Mut3-3
Mut3-4







Pro-activin A
63%
87%
84%
86%
89%


Other polypeptides
37%
13%
16%
14%
11%









The results indicate that degradation of the modified activin A of the present invention by the plant endogenous protease was significantly suppressed compared to the nonmodified activin A (N-His-Activin A, which is the wild type activin A) (FIG. 2).


This example demonstrated that the modified activin A of the present invention had the property of being hard to be degraded by a protease (resistance to degradation by a protease). This example also demonstrated that the modified activin A of the present invention was extremely useful in producing activin A in the presence of a protease, which is present in a host for expression, in a purification step, in a reagent and/or culture medium to be used, and the like. The reason for this is that the modified activin A of the present invention allows for improving the amount of expression thereof and/or the yield thereof compared to the nonmodified activin A.


2. Evaluation of Activity of Mature Activin A

The activity of the mature activin A obtained in “4. Production of Mature Activin A” of Example 2 was evaluated. F5-5 cells cultured in an incubator (37° C., 5% CO2) were seeded in a 96 well plate and cultured in the incubator after adding each mature activin A. Then, the differentiation rate of the cells was determined by staining the cells with AEC (aminoethylcarbazole, Tokyo Chemical Industry Co., Ltd.), according to the method described in Non Patent Literature 5 (Koretz K. et al., Histochemistry, 86: 5, 471-8, 1987). Table 2 shows the ED50 value of the mature activin A derived from the wild type activin A (N-His-Activin A) and the mature activin A derived from the modified activin A (Mut3-1 to Mut3-4).












TABLE 2







Expressed protein
ED50 ng/mL



















N-His-Activin A
<5



Mut3-1
<5



Mut3-2
<5



Mut3-3
<5



Mut3-4
<5










As shown in Table 2, the ED50 values of the mature activin A derived from the modified activin A (“Mut3-1” to “Mut3-4” in Table 2) were all less than 5 ng/mL, which were comparable to those of the mature activin A derived from the wild-type activin A (“N-His-Activin A” in Table 2).


These results indicated that the mature activin A obtained by treating the modified activin A with Furin had an ability to induce differentiation of the F5-5 cell into an erythroblast, with the same level of the wild-type activin A, and thus, it had an activity as mature activin A with same level of the activity of the wild-type activin A.


INDUSTRIAL APPLICABILITY

Use of the modified activin A of the present invention allows for improving the yield of activin A in producing the same in the presence of a protease.


[Sequence Listing Free Text]

SEQ ID NOs: 9 to 32 and 37: synthetic peptides


SEQ ID NOs: 33 to 36, 38, and 39: synthetic DNA

Claims
  • 1. Activin A having resistance to degradation by a protease other than Furin.
  • 2. The activin A according to claim 1, wherein the protease is an endogenous plant protease.
  • 3. The activin A according to claim 1, wherein the activin A comprises a modified proregion.
  • 4. The activin A according to claim 3, wherein the modified proregion comprises an amino acid sequence in which at least one amino acid is deleted, substituted and/or added in an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6.
  • 5. Activin A, comprising a modified proregion, wherein the modified proregion comprises an amino acid sequence in which at least one amino acid is deleted, substituted and/or added in an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6.
  • 6. The activin A according to claim 5, wherein the modified proregion comprises any amino acid sequence selected from the group consisting of (a) to (d): (a) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is deleted;(b) an amino acid sequence in which an amino acid sequence from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a spacer sequence consisting of 1 to 10 amino acids;(c) an amino acid sequence in which at least one of amino acids from position 182 to 199 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 is substituted with a different amino acid; and(d) an amino acid sequence in which an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6 has at least one amino acid inserted therein.
  • 7. The activin A according to claim 6, wherein the amino acid constituting the spacer sequence in the amino acid sequence (b) is at least one amino acid selected from the group consisting of glycine, alanine, and serine.
  • 8. The activin A according to claim 6, wherein the different amino acid in the amino acid sequence (c) is selected from the group consisting of alanine, serine, glycine, valine, leucine, and isoleucine.
  • 9. The activin A according to claim 5, wherein the activin A has resistance to degradation by a protease other than Furin.
  • 10. The activin A according to claim 5, wherein the modified proregion comprises any of amino acid sequences selected from the group consisting of (e) to (g): (e) an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20;(f) an amino acid sequence in which one or several amino acids are deleted, substituted or added in an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20, and activin A comprising the amino acid sequence has resistance to degradation by Furin; and(g) an amino acid sequence having 80% or more homology with an amino acid sequence set forth in any of SEQ ID NOs: 9 to 20, and activin A comprising the amino acid sequence has resistance to degradation by Furin.
  • 11. A polynucleotide encoding the activin A according to claim 1.
  • 12. A vector comprising the polynucleotide according to claim 11.
  • 13. A transformant expressing the activin A according to claim 1.
  • 14. The transformant according to claim 13, wherein the transformant is a plant transformant.
  • 15. The transformant according to claim 14, wherein the transformant expresses the activin A in apoplast.
  • 16. A method for producing activin A, the method comprising: recovering the activin A expressed in the transformant according to claim 13.
  • 17. A method for producing activin A, the method comprising: (a) recovering the activin A expressed in the transformant according to claim 13; and(b) treating the obtained activin A with Furin.
  • 18. Activin A that is a mixture of (i) activin A comprising a modified proregion and(ii) mature activin A,wherein the modified proregion comprises an amino acid sequence in which at least one amino acid is deleted, substituted and/or added in an amino acid sequence from position 180 to 201 of an amino acid sequence set forth in any of SEQ ID NOs: 4 to 6.
Priority Claims (1)
Number Date Country Kind
2018-214630 Nov 2018 JP national
Continuations (1)
Number Date Country
Parent PCT/JP2019/044744 Nov 2019 US
Child 17321049 US