CELL-PERMEABLE (CP)-CAS9 RECOMBINANT PROTEIN AND USES THEREOF

Information

  • Patent Application
  • 20190233835
  • Publication Number
    20190233835
  • Date Filed
    September 27, 2017
    6 years ago
  • Date Published
    August 01, 2019
    4 years ago
Abstract
The present invention relates to providing cell-permeable (CP)-Cas9 recombinant protein and uses thereof. Preferably, the CP-Cas9 recombinant protein may be used as Cas9 nuclease for CRISPR/Cas9 system by utilizing the platform technology for macromolecule intracellular transduction.
Description
TECHNICAL FIELD

The present invention relates to providing cell-permeable (CP)-Cas9 recombinant protein and uses thereof. Preferably, the CP-Cas9 recombinant protein may be used as Cas9 nuclease for CRISPR/Cas9 system by utilizing the platform technology for macromolecule intracellular transduction.


BACKGROUND ART

Epigenetics means over or above genetics that refers to hereditary changes in genome expression that do not involve alteration of DNA sequences. Epigenetics is a study for physiological phenotypic trait variations that are caused by external or environmental factors that switch genes on and off. Hence, improvement of epigenetic research relies on a wide range of gene editing technology.


The gene editing technology is the most powerful tool to insert, replace, and delete targeted DNA from genome. DNA sequence-specific recombination has been widely used for the gene editing technology to regulate genetic modifications, such as conditional gene expression, conditional mutagenesis, gene replacement and chromosome engineering in mammalian. There are several engineered nucleases being used: Transcription Activator-Like Effector Nucleases (TALENs), CRISPR/Cas9 system, Sleeping Beauty, and PiggyBac, Cre/LoxP system and Flp/Frt systems.


CRISPR/Cas system is originated from prokaryotic immune system. In the immune system, short segments of spacer sequence from previous exposures to a bacterial virus are inserted in the CRISPRs (Clustered Regularly Interspaced Short Palindromic Repeats). The spacer sequence is separated with short repetitions of base sequences and then recognizes foreign exogenous genetic elements, which is similar to the sequence of the spacer sequence. Cas9 nuclease cut the recognized foreign elements by complex with tracr RNA and crRNA (CRISPR RNA) consisting of spacer sequence and short repetitions.


CRISPR/Cas9 system is required for three components: Cas9, crRNA and tracr RNA. For genome editing, the crRNA and tracr RNA are combined to single guide RNA. Thus, the guide RNA (gRNA) binds to target sequence and then cleaves the sequence by Cas9 nuclease. To repair the breaks, the deleted sequence is silenced through mechanism mediated by NHEJ or the sequence is replaced by mechanism mediated by HDR using a donor template plasmid. The CRISPR/Cas9 system has been widely adopted. This has already been successfully used to target important genes in many cell lines and organisms, including human, bacteria, zebrafish, C. elegans, plants, Xenopus tropicalis, yeast, Drosophila, monkeys, rabbits, pigs, rats and mice. A recent exciting development is the use of the CRISPR/Cas9 system to target protein domains for transcriptional regulation, epigenetic modification, and microscopic visualization of specific genome loci.


REFERENCES

1. Fischer P M., Cellular uptake mechanisms and potential therapeutic utility of peptidic cell delivery vectors: progress 2001-2006, Med Res Rev. 2007;27:755-95.


2. Heitz F, Morris M C, Divita G., Twenty years of cell-penetrating peptides: from molecular mechanisms to therapeutics, Br J Pharmacol. 2009;157:195-206.


3. Lapenna S, Giordano A., Cell cycle kinases as therapeutic targets for cancer, Nat Rev Drug Discov. 2009;8:547-66.


4. Lim J, Kim J, Duong T, Lee G, Kim J, Yoon J. et al., Antitumor activity of cell-permeable p18(INK4c) with enhanced membrane and tissue penetration, Mol Ther. 2012;20:1540-9.


5. Jo D, Liu D, Yao S, Collins R D, Hawiger J., Intracellular protein therapy with SOCS3 inhibits inflammation and apoptosis, Nat Med. 2005;11:892-8.


6. Jo D, Nashabi A, Doxsee C, Lin Q, Unutmaz D, Chen J. et al., Epigenetic regulation of gene structure and function with a cell-permeable Cre recombinase, Nat Biotechnol. 2001;19:929-33.


7. Liu D, Li C, Chen Y, Burnett C, Liu X Y, Downs S. et al., Nuclear import of proinflammatory transcription factors is required for massive liver apoptosis induced by bacterial lipopolysaccharide, J Biol Chem. 2004;279:48434-42.


8. Liu D, Liu X Y, Robinson D, Burnett C, Jackson C, Seele L. et al., Suppression of Staphylococcal Enterotoxin B-induced Toxicity by a Nuclear Import Inhibitor, J Biol Chem. 2004;279:19239-46.


9. Liu D, Zienkiewicz J, DiGiandomenico A, Hawiger J., Suppression of acute lung inflammation by intracellular peptide delivery of a nuclear import inhibitor, Mol Ther. 2009;17:796-802.


10. Moore D J, Zienkiewicz J, Kendall P L, Liu D, Liu X, Veach R A. et al., In vivo islet protection by a nuclear import inhibitor in a mouse model of type 1 diabetes, PLoS One. 2010;5 :e13235.


11. Lim J, Jang G, Kang S, Lee G, Nga do T T, Phuong do T L. et al., Cell-.permeable NM23 blocks the maintenance and progression of established pulmonary metastasis, Cancer Res. 2011;71:7216-25.


12. Duong T, Kim J, Ruley H E, Jo D., Cell-permeable parkin proteins suppress Parkinson disease-associated phenotypes in cultured cells and animals, PLoS One. 2014;9:e102517.


13. Lim J, Duong T, Do N, Do P, Kim J, Kim H. et al., Antitumor activity of cell-permeable RUNX3 protein in gastric cancer cells. Clin Cancer Res. 2013;19:680-90.


14. Lim J, Duong T, Lee G, Seong B L, El-Rifai W, Ruley H E et al. The effect of intracellular protein delivery on the anti-tumor activity of recombinant human endostatin, Biomaterials. 2013;34:6261-71.


15. Lim J, Kim J, Kang J, Jo D., Partial somatic to stem cell transformations induced by cell-permeable reprogramming factors, Scientific Reports. 2014;4:4361.


DISCLOSURE
[Technical Problem]

The recombinant Cas9 protein that known in the art, has been used for CRISPR/Cas9 system. The recombinant Cas9 protein was not fused to macromolecule transduction domain, and displayed extremely low solubility, poor yields and relatively low cell- and tissue-permeability. Therefore, the recombinant Cas9 protein was not suitable for further CRISPR/Cas9 system development.


[Technical Solution]

For MITT, six critical factors (length, bending potential, instability index, aliphatic index, GRAVY, amino acid composition) have been determined through analysis of baseline hydrophobic CPPs. Advanced macromolecule transduction domain (aMTD), newly designed based on these six critical factors, could optimize cell-/tissue-permeability of Cas9 protein that has efficient gene editing and controlling an expression of target sequence. Further, in order to increase solubility and yield of recombinant protein, solubilization domains (SDs) additionally fused to the aMTD-Cas9 recombinant protein, thereby notably increased the solubility and manufacturing yield of the recombinant protein.


In this application, aMTD/SD-fused CP-Cas9 recombinant protein (CP-Cas9), much improved physicochemical characteristics (solubility and yield) and functional activity (cell-/tissue-permeability) compared with the recombinant Cas9 protein. In addition, the newly developed CP-Cas9 recombinant protein has now been demonstrated to insert a target sequence (new sequence) between original sequence, or delete a target sequence that bind with guide RNA and replace other sequence. The present application represents that macromolecule intracellular transduction technology (MITT) enabled by the new hydrophobic CPPs that are aMTD may provide novel protein to mediate conditional knockout of a target sequence or gene. These findings suggest that CP-Cas9 recombinant protein with improved cell/tissue-permeability completely suppress an expression or function of the target gene.


One aspect disclosed in the present application provides a Cell-Permeable (CP)-Cas9 recombinant protein, which comprises a Cas9 protein; and at least one advanced macromolecule transduction domain (aMTD) being composed of 9 to 13 amino acid sequences and having improved cell and/or tissue permeability, wherein the aMTD is fused to one end or both ends of the Cas9 protein and has the following features of:


(a) being composed of 3 or more amino acid sequences selected from the group consisting of Ala, Val, Ile, Leu, and Pro;


(b) having Proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and the last of its amino acid sequence; and


(c) having an instability index of 40 to 60; an aliphatic index of 180 to 220; and a grand average of hydropathy (GRAVY) of 2.1 to 2.6, as measured by Protparam.


According to one embodiment, the CP-Cas9 recombinant protein further comprises one or more solubilization domain (SD)(s), and the aMTD(s), Cas9 protein and SD(s) may be randomly fused to one another.


According to another embodiment, the aMTD may form a-Helix structure. According to still another embodiment, the aMTD may be composed of 12 amino acid sequences and represented by the following general formula:




embedded image


wherein X(s) independently refer to Alanine (A), Valine (V), Leucine (L) or Isoleucine (I); and Proline (P) can be positioned in one of U(s) (either 5′, 6′, 7′ or 8′). The remaining U(s) are independently composed of A, V, L or I, P at the 12′ is Proline.


Another aspect disclosed in the present application provides a CP-Cas9 recombinant protein which is represented by any one of the following structural formulae:


A-B-C, A-C-B, B-A-C, B-C-A, C-A-B, C-B-A, A-C-B-C and other possible combinations,


wherein A is an advanced macromolecule transduction domain (aMTD) having improved cell and/or tissue permeability, B is a Cas9 protein, and C is a solubilization domain (SD); and


the aMTD is composed of 9 to 13 amino acid sequences and has the following features of:


(a) being composed of 3 or more amino acids selected from the group consisting of Ala, Val, Ile, Leu, and Pro;


(b) having Proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and the last of its amino acid sequence;


(c) having an instability index of 40 to 60; an aliphatic index of 180 to 220; and a grand average of hydropathy (GRAVY) of 2.1 to 2.6, as measured by Protparam; and


(d) forming α-Helix structure.


According to one embodiment disclosed in the present application, the Cas9 protein may have an amino acid sequence of SEQ ID NO: 1221.


According to another embodiment disclosed in the present application, the Cas9 protein may be encoded by a polynucleotide sequence of SEQ ID NO: 1222.


According to still another embodiment disclosed in the present application, the Cas9 protein may further include a ligand selectively binding to a receptor of a cell, a tissue, or an organ.


According to still another embodiment disclosed in the present application, the at least one aMTD(s) may have an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 1 to 240.


According to still another embodiment disclosed in the present application, the at least one aMTD(s) may be encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 241 to 480.


According to still another embodiment disclosed in the present application, the one or more SD(s) may have an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 1203 to 1209.


According to still another embodiment disclosed in the present application, the one or more SD(s) may be encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 1210 to 1216.


According to still another embodiment disclosed in the present application, the CP-Cas9 recombinant protein may have a histidine-tag affinity domain additionally fused to one end thereof.


According to still another embodiment disclosed in the present application, the histidine-tag affinity domain may have an amino acid sequence of SEQ ID NO: 1217.


According to still another embodiment disclosed in the present application, the histidine-tag affinity domain may be encoded by a polynucleotide sequence of SEQ ID NO: 1218.


According to still another embodiment disclosed in the present application, the fusion may be formed via a peptide bond or a chemical bond.


Still another aspect disclosed in the present application provides a polynucleotide sequence encoding the CP-Cas9 recombinant protein.


Still another aspect disclosed in the present application provides a recombinant expression vector including the polynucleotide sequence.


Still another aspect disclosed in the present application provides a transformant transformed with the recombinant expression vector.


Still another aspect disclosed in the present application provides a preparing method of the CP-Cas9 recombinant protein including preparing the recombinant expression vector; preparing the transformant using the recombinant expression vector; culturing the transformant; and recovering the recombinant protein expressed by the culturing.


Still another aspect disclosed in the present application provides a composition including the CP-Cas9 recombinant protein as an active ingredient.


According to one embodiment disclosed in the present application, the composition may be used for insertion, replacement or deletion of a target sequence or gene.


Still another aspect disclosed in the present application provides the CP-Cas9 recombinant protein for insertion, replacement or deletion of a target sequence or gene.


Still another aspect disclosed in the present application provides use of the CP-Cas9 recombinant protein for insertion, replacement or deletion of a target sequence or gene.


Still another aspect disclosed in the present application provides a method of inserting, replacing or deleting of a target sequence or gene in a subject, the method including identifying a subject in need of inserting, replacing or deleting target sequence or gene; and administering to the subject an effective amount of the CP-Cas9 recombinant protein.


According to one embodiment disclosed in the present application, the subject may be a animal cell or plant cell.


Unless defined otherwise, all terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the present invention belongs. Although a certain method and a material is described herein, it should not be construed as being limited thereto, any similar or equivalent method and material to those may also be used in the practice or testing of the present invention. All publications mentioned herein are incorporated herein by reference in their entirety to disclose and describe the methods and/or materials in connection with which the publications are cited. It must be noted that as used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise.


A “peptide,” as used herein, refers to a chain-type polymer formed by amino acid residues which are linked to each other via peptide bonds, and used interchangeably with “polypeptide.” Further, a “polypeptide” includes a peptide and a protein.


Further, the term “peptide” includes amino acid sequences that are conservative variations of those peptides specifically exemplified herein. The term “conservative variation,” as used herein, denotes the replacement of an amino acid residue by another, biologically similar residue. Examples of conservative variations include substitution of one hydrophobic residue, such as isoleucine, valine, leucine, alanine, cysteine, glycine, phenylalanine, proline, tryptophan, tyrosine, nor leucine, or methionine for another, or substitution of one polar residue for another, for example, substitution of arginine for lysine, glutamic acid for aspartic acid, or glutamine for asparagine, and the like. Neutral hydrophilic amino acids which may be substituted for one another include asparagine, glutamine, serine, and threonine.


The term “conservative variation” also includes use of a substituted amino acid in place of an unsubstituted parent amino acid, provided that antibodies raised to the substituted polypeptide also immunoreact with the unsubstituted polypeptide. Such conservative substitutions are within the definition of the classes of the peptides disclosed in the present application.


A person having ordinary skill in the art may make similar substitutions to obtain peptides having higher cell permeability and a broader host range. For example, one aspect disclosed in the present application provides peptides corresponding to amino acid sequences (e.g. SEQ ID NOs: 1 to 240) provided herein, as well as analogues, homologs, isomers, derivatives, amidated variations, and conservative variations thereof, as long as the cell permeability of the peptide remains.


Minor modifications to primary amino acid sequence disclosed in the present application may result in peptides which have substantially equivalent or enhanced cell permeability, as compared to the specific peptides described herein. Such modifications may be deliberate, as by site-directed mutagenesis, or may be spontaneous.


All peptides may be synthesized using L-amino acids, but D forms of all of the peptides may be synthetically produced. In addition, C-terminal derivatives, such as C-terminal methyl esters and C-terminal amidates, may be produced in order to increase the cell permeability of the peptide according to one embodiment disclosed in the present application.


All of the peptides produced by these modifications are included herein, as long as in the case of amidated versions of the peptide, the cell permeability of the original peptide is altered or enhanced such that the amidated peptide is therapeutically useful. It is envisioned that such modifications are useful for altering or enhancing cell permeability of a particular peptide.


Furthermore, deletion of one or more amino acids may also result in a modification to the structure of the resultant molecule without any significant change in its cell permeability. This may lead to the development of a smaller active molecule which may also have utility. For example, amino- or carboxyl-terminal amino acids which may not be required for the cell permeability of a particular peptide may be removed.


The term “gene” refers to an arbitrary nucleic acid sequence or a part thereof having a functional role in protein coding or transcription, or regulation of other gene expression. The gene may be composed of all nucleic acids encoding a functional protein or a part of the nucleic acid encoding or expressing the protein. The nucleic acid sequence may include a gene mutation in exon, intron, initiation or termination region, promoter sequence, other regulatory sequence, or a unique sequence adjacent to the gene.


The term “primer” refers to an oligonucleotide sequence that hybridizes to a complementary RNA or DNA target polynucleotide and serves as the starting points for the stepwise synthesis of a polynucleotide from mononucleotides by the action of a nucleotidyltransferase as occurs, for example, in a polymerase chain reaction.


The term “coding region” or “coding sequence” refers to a nucleic acid sequence, a complement thereof, or a part thereof which encodes a particular gene product or a fragment thereof for which expression is desired, according to the normal base pairing and codon usage relationships. Coding sequences include exons in genomic DNA or immature primary RNA transcripts, which are joined together by the cellular biochemical machinery to provide a mature mRNA. The anti-sense strand is the complement of the nucleic acid, and the coding sequence may be deduced therefrom.


One aspect disclosed in the present application provides an CP-Cas9 recombinant protein, which comprises a Cas9 protein and at least one advanced macromolecule transduction domain (aMTD)(s) being composed of 9 to 13 amino acid sequences, preferably 10 to 12 amino acid sequences, and having improved cell and/or tissue permeability, wherein the aMTD is fused to one end or both ends of the Cas9 protein and has the following features of:


(a) being preferably composed of 3 or more amino acid sequences selected from the group consisting of Ala, Val, Ile, Leu, and Pro;


(b) having Proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and the last of its amino acid sequence, and preferably one or more of positions 5 to 8 and position 12 of its amino acid sequence; and


(c) having an instability index of preferably 40 to 60 and more preferably 41 to 58; an aliphatic index of preferably 180 to 220 and more preferably 185 to 225; and a grand average of hydropathy (GRAVY) of preferably 2.1 to 2.6 and more preferably 2.2 to 2.6 as measured by Protparam (see http://web.expasy.org/protparam/).


These critical factors that facilitate the cell permeable ability of aMTD sequences were analyzed, identified, and determined according to one embodiment disclosed in the present application. These aMTD sequences are artificially assembled based on the critical factors (CFs) determined from in-depth analysis of previously published hydrophobic CPPs.


The aMTD sequences according to one aspect disclosed in the present application are the first artificially developed cell permeable polypeptides capable of mediating the transduction of biologically active macromolecules—including peptides, polypeptides, protein domains, or full-length proteins—through the plasma membrane of cells.


According to one embodiment, the CP-Cas9 recombinant protein further comprises one or more solubilization domain (SD)(s), and the aMTD(s), Cas9 protein and SD(s) may be randomly fused to one another. For example, SD(s) may be further fused to one or more of the Cas9 protein and the aMTD, preferably one end or both ends of the Cas9 protein, and more preferably to the C-terminus of the Cas9 protein.


According to another embodiment, the aMTD may form a-Helix structure.


According to still another embodiment, the aMTD may be preferably composed of 12 amino acid sequences and represented by the following general formula:




embedded image


Here, X(s) independently refer to Alanine (A), Valine (V), Leucine (L) or Isoleucine (I); and Proline (P) can be positioned in one of U(s) (either 5′, 6′, 7′ or 8′). The remaining U(s) are independently composed of A, V, L or I, P at the 12′ is Proline.


Still another aspect disclosed in the present application provides an CP-Cas9 recombinant protein which is represented by any one of structural formulae A-B-C, A-C-B, B-A-C, B-C-A, C-A-B, C-B-A, A-C-B-C and other possible combinations, preferably by A-B-C or C-B-A:


wherein A is an advanced macromolecule transduction domain (aMTD) having improved cell and/or tissue permeability, and if the CP-Cas9 recombinant protein comprises two or more aMTDs, they can be same or different; B is a Cas9 protein; and C is a solubilization domain (SD), and if the CP-Cas9 recombinant protein comprises two or more SDs, they can be same or different; and


the aMTD is composed of 9 to 13, preferably 10 to 12 amino acid sequences and has the following features of:


(a) being composed of 3 or more amino acid sequences selected from the group consisting of Ala, Val, Ile, Leu, and Pro;


(b) having Proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and the last of its amino acid sequence, and preferably, one or more of positions 5 to 8 and position 12 of its amino acid sequence;


(c) having an instability index of 40 to 60, preferably 41 to 58 and more preferably 50 to 58; an aliphatic index of 180 to 220. preferably 185 to 225 and more preferably 195 to 205; and a grand average of hydropathy (GRAVY) of 2.1 to 2.6 and preferably 2.2 to 2.6, as measured by Protparam (see http://web.expasy.org/protparam/); and


(d) preferably forming a-Helix structure.


In one embodiment disclosed in the present application, the Cas9 protein may have an amino acid sequence of SEQ ID NO: 1221.


In another embodiment disclosed in the present application, the Cas9 protein may be encoded by a polynucleotide sequence of SEQ ID NO: 1222.


When the CP-Cas9 recombinant protein is intended to be delivered to a particular cell, tissue, or organ, the Cas9 protein may form a fusion product, together with an extracellular domain of a ligand capable of selectively binding to a receptor which is specifically expressed on the particular cell, tissue, or organ, or monoclonal antibody (mAb) capable of specifically binding to the receptor or the ligand and a modified form thereof.


The binding of the peptide and a biologically active substance may be formed either by indirect linkage by a cloning technique using an expression vector at a nucleotide level or by direct linkage via chemical or physical covalent or non-covalent bond of the peptide and the biologically active substance.


In still another embodiment disclosed in the present application, the Cas9 protein may preferably further include a ligand selectively binding to a receptor of a cell, a tissue, or an organ.


In one embodiment disclosed in the present application, the at least one aMTD(s) may have an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 1 to 240, preferably SEQ ID NOs: 39, 43, 63, 101, 121, 131, 147, 223 and 229, more preferably SEQ ID NO: 131.


In still another embodiment disclosed in the present application, the at least one aMTD(s) may be encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 241 to 480, preferably SEQ ID NOs: 279, 283, 303, 341, 361, 371, 387, 463 and 469, more preferably SEQ ID NO: 371.


In still another embodiment disclosed in the present application, the one or more SD(s) may have an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 1203 to 1209. The SD may be preferably SDA of SEQ ID NO: 1203, SDB of SEQ ID NO: 1204, or SDB' of SEQ ID NO: 1209, and more preferably, SDB of SEQ ID NO: 1204 which has superior structural stability, or SDB' of SEQ ID NO: 1209 which has a modified amino acid sequence of SDB to avoid immune responses upon in vivo application. The modification of the amino acid sequence in SDB may be replacement of an amino acid residue, Valine, corresponding to position 28 of the amino acid sequence of SDB (SEQ ID NO: 1204) by Leucine.


In still another embodiment disclosed in the present application, the one or more SDs may be encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 1210 to 1216. The SD may be preferably SDA encoded by a polynucleotide sequence of SEQ ID NO: 1210, SDB encoded by a polynucleotide sequence of SEQ ID NO: 1211, or SDB' for deimmunization (or humanization) encoded by a polynucleotide sequence of SEQ ID NO: 1216, and more preferably, SDB having superior structural stability, which is encoded by a polynucleotide sequence of SEQ ID NO: 1211, or SDB′ having a modified polynucleotide sequence of SDB to avoid immune responses upon in vivo application, which is encoded by a polynucleotide sequence of SEQ ID NO: 1216.


In still another embodiment disclosed in the present application, the CP-Cas9 recombinant protein may be preferably selected from the group consisting of:


1) a recombinant protein, in which Cas9 having an amino acid sequence of SEQ ID NO: 1221 is fused to the N-terminus or the C-terminus of aMTD having any one amino acid sequence selected from the group consisting of SEQ ID NOs: 1 to 240, more preferably SEQ ID NO: 1232;


2) a recombinant protein, in which SD having any one amino acid sequence selected from the group consisting of SEQ ID NOs: 1203 to 1209 is further fused to one or more of the N-terminus or the C-terminus of the Cas9 and aMTD in the recombinant protein of 1); and


3) a recombinant protein, in which a Histidine tag having an amino acid sequence of SEQ ID NOs: 1217 is further fused to the N-terminus of the recombinant protein of 1) or 2).


According to one embodiment, the CP-Cas9 recombinant protein may be composed of an amino acid sequence represented by SEQ ID NO: 1230.


The Cas9 protein may exhibit a physiological phenomenon-related activity or a therapeutic purpose-related activity by intracellular or in-vivo delivery. The recombinant expression vector may include a tag sequence which makes it easy to purify the recombinant protein, for example, consecutive histidine codon, maltose binding protein codon, Myc codon, etc., and further include a fusion partner to enhance solubility of the recombinant protein, etc. Further, for the overall structural and functional stability of the recombinant protein or flexibility of the proteins encoded by respective genes, the recombinant expression vector may further include one or more glycine, proline, and spacer amino acid or polynucleotide sequences including AAY amino acids. Furthermore, the recombinant expression vector may include a sequence specifically digested by an enzyme in order to remove an unnecessary region of the recombinant protein, an expression regulatory sequence, and a marker or reporter gene sequence to verify intracellular delivery, but is not limited thereto.


In still another embodiment disclosed in the present application, the CP-Cas9 recombinant protein may preferably have a histidine-tag affinity domain additionally fused to one end thereof.


In still another embodiment disclosed in the present application, the histidine-tag affinity domain may have an amino acid sequence of SEQ ID NO: 1217.


In still another embodiment disclosed in the present application, the histidine-tag affinity domain may be encoded by a polynucleotide sequence of SEQ ID NO: 1218.


In still another embodiment disclosed in the present application, the fusion may be formed via a peptide bond or a chemical bond.


The chemical bond may be preferably selected from the group consisting of disulfide bonds, diamine bonds, sulfide-amine bonds, carboxyl-amine bonds, ester bonds, and covalent bonds.


In still another embodiment disclosed in the present application, the CP-Cas9 recombinant protein may be used for insertion, replacement or deletion of a target sequence or gene.


Still another aspect disclosed in the present application provides a polynucleotide sequence encoding the CP-Cas9 recombinant protein.


According to still another embodiment disclosed in the present application, the polynucleotide sequence may be fused with a histidine-tag affinity domain.


Still another aspect disclosed in the present application provides a recombinant expression vector including the polynucleotide sequence.


Preferably, the vector may be inserted in a host cell and recombined with the host cell genome, or refers to any nucleic acid including a nucleotide sequence competent to replicate spontaneously as an episome. Such a vector may include a linear nucleic acid, a plasmid, a phagemid, a cosmid, an RNA vector, a viral vector, etc.


Preferably, the vector may be genetically engineered to incorporate the nucleic acid sequence encoding the recombinant protein in an orientation either N-terminal and/or C-terminal to a nucleic acid sequence encoding a peptide, a polypeptide, a protein domain, or a full-length protein of interest, and in the correct reading frame so that the recombinant protein consisting of aMTD, Cas9 protein, and preferably SD may be expressed. Expression vectors may be selected from those readily available for use in prokaryotic or eukaryotic expression systems.


Standard recombinant nucleic acid methods may be used to express a genetically engineered recombinant protein. The nucleic acid sequence encoding the recombinant protein according to one embodiment disclosed in the present application may be cloned into a nucleic acid expression vector, e.g., with appropriate signal and processing sequences and regulatory sequences for transcription and translation, and the protein may be synthesized using automated organic synthetic methods. Synthetic methods of producing proteins are described in, for example, the literature [Methods in Enzymology, Volume 289: Solid-Phase Peptide Synthesis by Gregg B. Fields (Editor), Sidney P. Colowick, Melvin I. Simon (Editor), Academic Press (1997)].


In order to obtain high level expression of a cloned gene or nucleic acid, for example, a cDNA encoding the recombinant protein according to one embodiment disclosed in the present application, the recombinant protein sequence may be typically subcloned into an expression vector that includes a strong promoter for directing transcription, a transcription/translation terminator, and in the case of a nucleic acid encoding a protein, a ribosome binding site for translational initiation. Suitable bacterial promoters are well known in the art and are described, e.g., in the literatures [Sambrook & Russell, Molecular Cloning: A Laboratory Manual, 3d Edition, Cold Spring Harbor Laboratory, N.Y. (2001); and Ausubel, et al., Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, N. Y. (1989)]. Bacterial expression systems for expression of the recombinant protein are available in, e.g., E. coli, Bacillus sp., and Salmonella (Palva et al., Gene 22: 229-235 (1983); Mosbach et al., Nature 302: 543-545 (1983)). Kits for such expression systems are commercially available. Eukaryotic expression systems for mammalian cells, yeast, and insect cells are well known in the art and are also commercially available. The eukaryotic expression vector may be preferably an adenoviral vector, an adeno-associated vector, or a retroviral vector.


Generally, the expression vector for expressing the cell permeable recombinant protein according to one embodiment disclosed in the present application in which the cargo protein, i.e. Cas9 protein, is attached to the N-terminus, C-terminus, or both termini of aMTD may include regulatory sequences including, for example, a promoter, operably attached to a sequence encoding the advanced macromolecule transduction domain. Non-limiting examples of inducible promoters that may be used include steroid-hormone responsive promoters (e.g., ecdysone-responsive, estrogen-responsive, and glutacorticoid-responsive promoters), tetracycline “Tet-On” and “Tet-Off” systems, and metal-responsive promoters.


The polynucleotide sequence according to one embodiment disclosed in the present application may be present in a vector in which the polynucleotide sequence is operably linked to regulatory sequences capable of providing for the expression of the polynucleotide sequence by a suitable host cell.


According to one embodiment disclosed in the present application, the polynucleotide sequence may be selected from the following groups:


1) a polynucleotide sequence, in which any one polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241 to 480, preferably SEQ ID NOs: 279, 283, 303, 341, 361, 371, 387, 463 and 469, more preferably SEQ ID NO: 371, is operably linked with a polynucleotide sequence of SEQ ID NO: 1233; and


2) a polynucleotide sequence, in which any one polynucleotide sequence selected from the group consisting of SEQ ID NOs: 1210 to 1216 is further operably linked to the polynucleotide sequence of 1), or further operably linked to between: any one polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241 to 480, preferably SEQ ID NOs: 279, 283, 303, 341, 361, 371, 387, 463 and 469, more preferably SEQ ID NO: 371; and a polynucleotide sequence of SEQ ID NO: 1231.


Within an expression vector, the term “operably linked” is intended to mean that the polynucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the polynucleotide sequence. The term “regulatory sequence” is intended to include promoters, enhancers, and other expression control elements. Such operable linkage with the expression vector can be achieved by conventional gene recombination techniques known in the art, while site-directed DNA cleavage and linkage are carried out by using conventional enzymes known in the art.


The expression vectors may contain a signal sequence or a leader sequence for membrane targeting or secretion, as well as regulatory sequences such as a promoter, an operator, an initiation codon, a termination codon, a polyadenylation signal, an enhancer and the like. The promoter may be a constitutive or an inducible promoter. Further, the expression vector may include one or more selectable marker genes for selecting the host cell containing the expression vector, and may further include a polynucleotide sequence that enables the vector to replicate in the host cell in question.


The expression vector constructed according to one embodiment disclosed in the present application may be the vector where the polynucleotide encoding the CP-Cas9 recombinant protein (where an aMTD is fused to the N-terminus or C-terminus of a Cas9 protein) is inserted within the multiple cloning sites (MCS), preferably within the Nde1/Sal1 site or BamH1/Sal1 site of a pET-28a(+)(Novagen, Darmstadt, Germany) or pET-26b(+) vector(Novagen, Darmstadt, Germany).


In still another embodiment disclosed in the present application, the polynucleotide encoding the SD being additionally fused to the N-terminus or C-terminus of a Cas9 protein or an aMTD may be inserted into a cleavage site of restriction enzyme (Nde1, BamH1 and Sal1, etc.) within the multiple cloning sites (MCS) of a pET-28a(+)(Novagen, Darmstadt, Germany) or pET-26b(+) vector(Novagen, Darmstadt, Germany).


In still another embodiment disclosed in the present application, the polynucleotide encoding the CP-Cas9 recombinant protein may be cloned into a pET-28a(+) vector bearing a His-tag sequence so as to fuse six histidine residues to the N-terminus of the CP-Cas9 recombinant protein to allow easy purification.


According to one embodiment disclosed in the present application, the polynucleotide sequence may be a polynucleotide sequence selected from the group consisting of SEQ ID NOs: 1231 and 1233.


The recombinant protein may be introduced into an appropriate host cell, e.g., a bacterial cell, a yeast cell, an insect cell, or a tissue culture cell. The recombinant protein may also be introduced into embryonic stem cells in order to generate a transgenic organism. Large numbers of suitable vectors and promoters are known to those skilled in the art and are commercially available for generating the recombinant protein.


Known methods may be used to construct vectors including the polynucleotide sequence according to one embodiment disclosed in the present application and appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo recombination/genetic recombination. For example, these techniques are described in the literatures [Sambrook & Russell, Molecular Cloning: A Laboratory Manual, 3d Edition, Cold Spring Harbor Laboratory, N. Y. (2001); and Ausubel et al., Current Protocols in Molecular Biology Greene Publishing Associates and Wiley Interscience, N.Y. (1989)].


Still another aspect disclosed in the present application provides a transformant transformed with the recombinant expression vector.


The transformation includes transfection, and refers to a process whereby a foreign (extracellular) DNA, with or without an accompanying material, enters into a host cell. The “transfected cell” refers to a cell into which the foreign DNA is introduced into the cell, and thus the cell harbors the foreign DNA. The DNA may be introduced into the cell so that a nucleic acid thereof may be integrated into the chromosome or replicable as an extrachromosomal element. The cell introduced with the foreign DNA, etc. is called a transformant.


As used herein, ‘introducing’ of a protein, a peptide, an organic compound into a cell may be used interchangeably with the expression of ‘carrying,’ ‘penetrating,’ ‘transporting,’ ‘delivering,’ ‘permeating’ or ‘passing.’


It is understood that the host cell refers to a eukaryotic or prokaryotic cell into which one or more DNAs or vectors are introduced, and refers not only to the particular subject cell but also to the progeny or potential progeny thereof. 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 as used herein.


The host cells may be preferably bacterial cells, and as the bacterial cells, there are, in principle, no limitations. They may be eubacteria (gram-positive or gram-negative) or archaebacteria, as long as they allow genetic manipulation for insertion of a gene of interest, preferably for site-specific integration, and they may be cultured on a manufacturing scale. Preferably, the host cells may have the property to allow cultivation to high cell densities.


Examples of bacterial host cells that may be used in the preparation of the recombinant protein are E. coli (Lee, 1996; Hannig and Makrides, 1998), Bacillus subtilis, Pseudomonas fluorescens (Squires et al., 2004; Retallack et al., 2006) as well as various Corynebacterium (US 2006/0003404 A1) and Lactococcus lactis (Mierau et al., 2005) strains. Preferably, the host cells are Escherichia coli cells.


More preferably, the host cell may include an RNA polymerase capable of binding to a promoter regulating the gene of interest. The RNA polymerase may be endogenous or exogenous to the host cell.


Preferably, host cells with a foreign strong RNA polymerase may be used. For example, Escherichia coli strains engineered to carry a foreign RNA polymerase (e.g. like in the case of using a T7 promoter a T7-like RNA polymerase in the so-called “T7 strains”) integrated in their genome may be used. Examples of T7 strains, e.g. BL21(DE3), HMS174(DE3), and their derivatives or relatives (see Novagen, pET System manual, 11th edition), may be widely used and commercially available. Preferably, BL21-CodonPlus (DE3)-RIL or BL21-CodonPlus (DE3)-RIPL (Agilent Technologies) may be used. These strains are DE3 lysogens containing the T7 RNA polymerase gene under control of the lacUV5 promoter. Induction with IPTG allows production of T7 RNA polymerase which then directs the expression of the gene of interest under the control of the T7 promoter.


The host cell strains, E. coli BL21(DE3) or HMS174(DE3), which have received their genome-based T7 RNA polymerase via the phage DE3, are lysogenic. It is preferred that the T7 RNA polymerase contained in the host cell has been integrated by a method which avoids, or preferably excludes, the insertion of residual phage sequences in the host cell genome since lysogenic strains have the disadvantage to potentially exhibit lytic properties, leading to undesirable phage release and cell lysis.


Still another aspect disclosed in the present application provides a preparing method of the CP-Cas9 recombinant protein including preparing the recombinant expression vector; preparing the transformant using the recombinant expression vector; culturing the transformant; and recovering the recombinant protein expressed by culturing.


Culturing may be preferably in a mode that employs the addition of a feed medium, this mode being selected from the fed-batch mode, semi-continuous mode, or continuous mode, and the bacterial expression host cells may include a DNA construct, integrated in their genome, carrying the DNA sequence encoding the protein of interest under the control of a promoter that enables expression of said protein.


There are no limitations in the type of the culture medium. The culture medium may be semi-defined, i.e. containing complex media compounds (e.g. yeast extract, soy peptone, casamino acids), or it may be chemically defined, without any complex compounds. Preferably, a defined medium may be used. The defined media (also called minimal or synthetic media) are exclusively composed of chemically defined substances, i.e. carbon sources such as glucose or glycerol, salts, vitamins, and, in view of a possible strain auxotrophy, specific amino acids or other substances such as thiamine. Most preferably, glucose may be used as a carbon source. Usually, the carbon source of the feed medium serves as the growth-limiting component which controls the specific growth rate.


Host cells may be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or the use of cell lysing agents. The literature [Scopes, Protein Purification: Principles and Practice, New York: Springer-Verlag (1994)] describes a number of general methods for purifying recombinant (and non-recombinant) proteins. The methods may include, e.g., ion-exchange chromatography, size-exclusion chromatography, affinity chromatography, selective precipitation, dialysis, and hydrophobic interaction chromatography. These methods may be adapted to devise a purification strategy for the cell permeable recombinant protein. If the cell permeable recombinant protein includes a purification handle, such as an epitope tag or a metal chelating sequence, affinity chromatography may be used to easily purify the protein.


The amount of the protein produced may be evaluated by detecting the advanced macromolecule transduction domain directly (e.g., using Western analysis) or indirectly (e.g., by assaying materials derived from the cells for specific DNA binding activity, such as by electrophoretic mobility shift assay). Proteins may be detected prior to purification, during any stage of purification, or after purification. In some implementations, purification or complete purification may not be necessary.


In one embodiment disclosed in the present invention, the CP-Cas9 recombinant protein may be used in a gene editing technology or chromatin engineering that recognizes a specific nucleotide sequence, cuts and edits the recognized sequence, and insert or replace other gene. The CP-Cas9 recombinant protein is capable of inducing a insertion of a specific gene(s) or a deletion of a specific gene(s) without modification of genome. The CP-Cas9 recombinant protein may recognize and exactly delete a target gene(s) (a mutant gene(s)) in whole or in part. More specifically, the CP-Cas9 recombinant protein recognizes a guide RNA (gRNA) binding to the target sequence or gene. Thus, it may be possible to cut a desired target sequence(s) by changing the guide RNA.


The CP-Cas9 recombinant protein may be applicable to both animal cell and plant cell. According to one embodiment disclosed in the present invention, the CP-Cas9 recombinant protein may be utilized to delete a target gene(s) from a fertilized egg or a stem cell, replace a normal gene(s) into a fertilized egg or a stem cell, thereby treating congenital hereditary diseases caused by a gene mutation. The CP-Cas9 recombinant protein may also be used to produce genetically modified organism (GMO) foods by introducing an insect-resistance gene(s) or a herbicide-resistance gene(s) into plant cells.


Still another aspect disclosed in the present application provides a composition including the CP-Cas9 recombinant protein as an active ingredient.


According to one embodiment disclosed in the present application, the composition may be used for insertion, replacement or deletion of a target sequence or gene.


The composition may preferably comprise the active ingredient in an amount of 0.1 to 99.9% by weight, based on the total weight of the composition. In addition to the above active ingredient, the composition may comprise a buffer, an ajuvant, etc. which is physiologically acceptable while stabilizing the recombinant protein.


Still another aspect disclosed in the present application provides the CP-Cas9 recombinant protein for the insertion, replacement or deletion of a target sequence or gene.


Still another aspect disclosed in the present application provides use of the CP-Cas9 recombinant protein for the insertion, replacement or deletion of a target sequence or gene.


Still another aspect disclosed in the present application provides a method of inserting, replacing or deleting target sequences in a subject including identifying a subject in need of inserting, replacing or deleting target sequence; and administering to the subject an effective amount of the CP-Cas9 recombinant protein.


In one embodiment disclosed in the present application, the subject may be preferably an animal cell or plant cell. More preferably, the animal cell may be fertilized egg or stem cell.


In one embodiment disclosed in the present invention, the CP-Cas9 recombinant protein, may be used to remove a mutant gene(s) by treating a fertilized egg including the mutant gene(s), which causes a hereditary disease, with the CP-Cas9 recombinant protein and a guide RNA recognizing the mutant gene(s). It may be also possible to insert a normal gene at the position of the removed gene(s) by using the CP-Cas9 recombinant protein. The treated fertilized egg may develop into a baby with no hereditary disease.


In another embodiment disclosed in the present invention, it may be possible to remove a gene(s) inhibiting the growth of microorganisms with the CP-Cas9 recombinant protein, thereby inducing improvement of the microorganisms to be easily cultured and produced in large amounts.


[Advantageous Effects]

According to one aspect disclosed in the present application, cell-permeable Cas9 recombinant protein may effectively insert the sequence of target gene, or delete the sequence of target gene and replace other gene, and use for CRISPR/Cas9 system. The CRISPR/Cas9 system using the CP-Cas9 recombinant protein may be utilized to study of gene editing to insert new gene, or delete a mutant gene and replace a mutant gene to normal gene.


However, the effects of the disclosures in the present application are not limited to the above-mentioned effects, and another effects not mentioned will be clearly understood by those skilled in the art from the following description.





DESCRIPTION OF DRAWINGS


FIG. 1 shows structures of aMTD- or rPeptide-fused recombinant proteins. A schematic diagram of the His-tagged CRA recombinant proteins is illustrated and constructed according to the present invention. The his-tag for affinity purification (white), aMTD or rPeptide (gray) and cargo A (CRA, black) are shown.



FIG. 2a shows construction of expression vectors for aMTDs- or rPeptide-fused recombinant proteins. FIGS. 2b and 2c show the agarose gel electrophoresis analysis showing plasmid DNA fragments at 645 bp insert encoding aMTDs or rPeptide-fused CRA cloned into the pET28a(+) vector according to the present invention.



FIGS. 3a to 3d show inducible expression of aMTD- or rPeptide-fused recombinant proteins. Expressed recombinant aMTD- or random peptide-fused CRA recombinant proteins were transformed in E. coli BL21 (DE3) strain. Expression of recombinant proteins in E.coli before (−) and after (+) induction with IPTG was monitored by SDS-PAGE, and stained with Coomassie blue.



FIGS. 4a and 4b show purification of aMTD- or rPeptide-fused recombinant proteins. Expressed recombinant proteins were purified by Ni2+ affinity chromatography under the natural condition. Purification of recombinant proteins displayed through SDS-PAGE analysis.



FIGS. 5a to 5u show determination of aMTD-mediated cell-permeability. Cell-permeability of a negative control (A: rP38) and reference hydrophobic CPPs (MTM12 and MTD85) are shown. The cell-permeability of each aMTD and/or rPeptide is visually compared to that of the cargo protein lacking peptide sequence (HCA). Gray shaded area represents untreated RAW 264.7 cells (vehicle); thin light gray line represents the cells treated with equal molar concentration of FITC (FITC only); dark thick line indicates the cells treated with FITC-his-tagged CRA protein (HCA); and the cells treated with the FITC-proteins (HMCA) fused to negative control (rP38), reference CPP (MTM12 or MTD85) or new hydrophobic CPP (aMTD) are shown with light thick line and indicated by arrows.



FIGS. 6a to 6c show determination of rPeptide-mediated cell-permeability. The cell-permeability of each aMTD and/or rPeptide was visually compared to that of the cargo protein lacking peptide sequence (HCA). Gray shaded area represents untreated RAW 264.7 cells (vehicle); thin light gray line represents the cells treated with equal molar concentration of FITC (FITC only); dark thick line indicates the cells treated with FITC-his-tagged CRA protein (HCA); and the cells treated with the FITC-proteins fused to rPeptides are shown with light thick line and indicated by arrows.



FIGS. 7a to 7k show visualized cell-permeability of aMTD-fused recombinant proteins. NIH3T3 clls were treated with FITC-labeled protein (10 μM) fused to aMTD for 1 hour at 37. Cell-permeability of the proteins was visualized by laser scanning confocal microscopy (LSM700 version).



FIG. 8 shows visualized cell-permeability of rPeptide-fused recombinant proteins. Cell-permeability of rPeptide-fused recombinant proteins was visualized by laser scanning confocal microscopy (LSM700 version).



FIGS. 9a to 9c show relative cell-permeability of aMTD-fused recombinant proteins compared to negative control (rP38). The FIG shows graphs comparing the cell-permeability of the recombinant proteins fused to aMTDs and a negative control (A: rP38).



FIGS. 10a to 10c show relative cell-permeability of aMTD-fused recombinant proteins compared to reference CPP (MTM12). The FIG shows graphs comparing the cell-permeability of the recombinant proteins fused to aMTDs and a reference CPP (MTM12).



FIGS. 11a to 11c show relative cell-permeability of aMTD-fused recombinant proteins compared to reference CPP (MTD85). The FIG shows graphs comparing the cell-permeability of the recombinant proteins fused to aMTDs and a reference CPP (MTD85).



FIG. 12 shows relative cell-permeability of rPeptide-mediated recombinant proteins compared to average that of aMTDs. The FIG shows graphs comparing the cell-permeability of the recombinant proteins fused to rPeptides and that (average value: aMTD AVE) of aMTDs.



FIGS. 13a to 13d show association of cell-permeability with amino acid composition in aMTD sequences. These graphs display delivery potential (Geometric Mean) of aMTDs influenced with amino acid composition (A, I, V and L).



FIGS. 14a and 14b show association of cell-permeability with critical factors in aMTDs. These graphs show the association of cell-permeability with critical factors [bending potential: proline position (PP), rigidity/flexibility: instability index (II), structural feature: aliphatic index (AI) and hydropathy: grand average of hydropathy (GRAVY)].



FIGS. 15a and 15b show relative relevance of aMTD-mediated cell-permeability with critical factors. Cell-permeability of 10 high and 10 low ranked aMTDs in their delivery potential were examined for their association with the critical factors [bending potential: proline position (PP), rigidity/flexibility: instability index (II), structural feature: aliphatic index (AI) and hydropathy: grand average of hydropathy (GRAVY)].



FIG. 16 shows relative relevance of rPeptide-mediated cell-permeability with hydropathy range (GRAVY). This graph and a chart illustrate relative relevance of rPeptide-mediated cell-permeability with its hydropathy range (GRAVY).



FIG. 17 shows structures of Cas9 recombinant protein designed according to example 6-1.



FIG. 18 shows an agarose gel electrophoresis analysis showing plasmid DNA fragments insert encoding His-Cas9 (HC9) or His-NLS-aMTD563-Cas9-SDB (HNM563C9SB) cloned into the pET28a (+) vector according to example 6-1.



FIG. 19 shows inducible expression and purification of CP-Cas9 recombinant protein in E.coli according to example 6-2 and improvement of solubility/yield of CP-Cas9 recombinant protein by fusing aMTD/SD according to example 6-3.



FIG. 20 shows structures of Cas9 recombinant protein fused various aMTDs and SDB.



FIG. 21 shows aMTD-mediated cell-permeability of CP-Cas9 recombinant protein in RAW 264.7 cells according to example 7-1.



FIG. 22 shows aMTD-mediated intracellular delivery and localization of CP-Cas9 recombinant protein in HeLa cells cells according to example 7-1.



FIG. 23 shows nuclease activity of CP-Cas9 recombinant protein according to example 8-1.



FIG. 24 shows nuclease activity of CP-Cas9 recombinant protein (in dose dependent manner of gRNA) according to example 8-2.



FIG. 25 shows nuclease activity of CP-Cas9 recombinant protein (in dose dependent manner of CP-Cas9) according to example 8-3.



FIG. 26 shows nuclease activity of CP-Cas9 recombinant protein according to example 8-4.



FIG. 27 shows a structure of the RFP plasmid according to example 9-1 to 9-3.



FIG. 28 shows biological activity of CP-Cas9 recombinant protein according to example 9-1.



FIG. 29 shows biological activity of CP-Cas9 recombinant protein according to example 9-2.



FIG. 30 shows biological activity of CP-Cas9 recombinant protein according to example 9-3.





MODE FOR INVENTION
1. Analysis of Reference Hydrophobic CPPs to Identify ‘Critical Factors’ for Development of Advanced MTDs

Previously reported MTDs were selected from a screen of more than 1,500 signal peptide sequences. Although the MTDs that have been developed did not have a common sequence or sequence motif, they were all derived from the hydrophobic (H) regions of signal sequences (HRSSs) that also lack common sequences or motifs except their hydrophobicity and the tendency to adopt alpha-helical conformations. The wide variation in H-region sequences may reflect prior evolution for proteins with membrane translocating activity and subsequent adaptation to the SRP/Sec61 machinery, which utilizes a methionine-rich signal peptide binding pocket in SRP to accommodate a wide-variety of signal peptide sequences.


Previously described hydrophobic CPPs (e.g. MTS/MTM and MTD) were derived from the hydrophobic regions present in the signal peptides of secreted and cell surface proteins. The prior art consists first, of ad hoc use of H-region sequences (MTS/MTM), and second, of H-region sequences (with and without modification) with highest CPP activity selected from a screen of 1,500 signal sequences (MTM). Second prior art, the modified H-region derived hydrophobic CPP sequences had advanced in diversity with multiple number of available sequences apart from MTS/MTM derived from fibroblast growth factor (FGF) 4. However, the number of MTDs that could be modified from naturally occurring secreted proteins are somewhat limited. Because there is no set of rules in determining their cell-permeability, no prediction for the cell-permeability of modified MTD sequences can be made before testing them.


The hydrophobic CPPs, like the signal peptides from which they originated, did not conform to a consensus sequence, and they had adverse effects on protein solubility when incorporated into protein cargo. We therefore set out to identify optimal sequence and structural determinants, namely critical factors (CFs), to design new hydrophobic CPPs with enhanced ability to deliver macromolecule cargoes including proteins into the cells and tissues while maintaining protein solubility. These newly developed CPPs, advanced macromolecule transduction domains (aMTDs) allowed almost infinite number of possible designs that could be designed and developed based on the critical factors. Also, their cell-permeability could be predicted by their character analysis before conducting any in vitro and/or in vivo experiments. These critical factors below have been developed by analyzing all published reference hydrophobic CPPs.


1-1. Analysis of Hydrophobic CPPs

Seventeen different hydrophobic CPPs (Table 1) published from 1995 to 2014 (Table 2) were selected. After physiological and chemical properties of selected hydrophobic CPPs were analyzed, 11 different characteristics that may be associated with cell-permeability have been chosen for further analysis. These 11 characteristics are as follows: sequence, amino acid length, molecular weight, pI value, bending potential, rigidity/flexibility, structural feature, hydropathy, residue structure, amino acid composition and secondary structure of the sequences (Tables 3 and 4).


Table 1 shows the summary of published hydrophobic Cell-Penetrating Peptides which were chosen.













TABLE 1





#
Pepides
Origin
Protein
Ref.



















1
MTM

Homo sapiens

NP_001998 Kaposi fibroblast growth factor (K-FGF)
1


2
MTS

Homo sapiens

NP_001998 Kaposi fibroblast growth factor (K-FGF)
2


3
MTD10

Streptomyces coelicolor

NP_625021 Glycosyl hydrolase
8


4
MTD13

Streptomyces coelicolor

NP_639877 Putative secreted protein
3


5
MTD47

Streptomyces coelicolor

NP_627512 Secreted protein
4


6
MTD56

Homo sapiens

P23274 Peptidyl-prolyl cis-trans isomerase B
5





precursor



7
MTD73

Drosophila melanogaster

AAA17887 Spatzle (spz) protein
5


8
MTD77

Homo sapiens

NP_003231 Kaposi fibroblast growth factor (K-FGF)
6


9
MTD84

Phytophthora cactorum

AAK63068 Phytotoxic protein PcF precusor
4


10
MTD85

Streptomyces coelicolor

NP_629842 Peptide transport system peptide binding
7





protein



11
MTD86

Streptomyces coelicolor

NP_629842 Peptide transport system secreted peptide
7





binding protein



12
MTD103

Homo sapiens

TMBV19 domain Family member B
8


13
MTD132

Streptomyces coelicolor

NP_628377 P60-family secreted protein
4


14
MTD151

Streptomyces coelicolor

NP_630126 Secreted chitinase
8


15
MTD173

Streptomyces coelicolor

NP_624384 Secreted protein
4


16
MTD174

Streptomyces coelicolor

NP_733505 Large, multifunctional secreted protein
8


17
MTD181

Neisseria meningitidis Z2491

CAB84257.1 Putative secreted protein
4









Table 2 summarizes reference information.











TABLE 2









References













#
Title
Journal
Year
Vol
Issue
Page
















1
Inhibition of Nuclear Translocation of
JOURNAL OF
1995
270
24
14255



Transcription Factor NF-kB by a Synthetic
BIOLOGICAL



peptide Containing a Cell
CHEMISTRY



Membrane-permeable Motif and Nuclear



Localization Sequence


2
Epigenetic Regulation of Gene Structure and
NATURE
2001
19
10
929



Function with a Cell-Permeable Cre
BIOTECHNOLOGY



Recombinase


3
Cell-Permeable NM23 Blocks the Maintenance
CANCER
2011
71
23
7216



and Progression of Established Pulmonary
RESEARCH



Metastasis


4
Antitumor Activity of Cell-Permeable
MOLECULAR
2012
20
8
1540



p18INK4c With Enhanced Membrane and
THERAPY



Tissue Penetration


5
Antitumor Activity of Cell-Permeable RUNX3
CLINICAL
2012
19
3
680



Protein in Gastric Cancer Cells
CANCER




RESEARCH


6
The Effect of Intracellular Protein Delivery on
BIOMATERIALS
2013
34
26
6261



the Anti-Tumor Activity of Recombinant



Human Endostatin


7
Partial Somatic to Stem Cell Transformations
SCIENTIFIC
2014
4
10
4361



Induced By Cell-Permeable Reprogramming
REPORTS



Factors


8
Cell-Permeable Parkin Proteins Suppress
PLOS ONE
2014
9
7
17



Parkinson Disease-Associated Phenotypes in



Cultured Cells and Animals









Tables 3 and 4 show characteristics of published hydrophobic Cell-Penetrating Peptides (A) which were analyzed (SEQ ID NOs: 798 to 814).


















TABLE 3













Structural










Rigidity/Flexibility
Feature






Molecular

Bending
(Instability
(Aliphatic
Hydropathy


#
Peptides
Sequence
Length
Weight
pI
Potential
Index: II)
Index: AI)
(GRAVY)
























1
MTM
AAVALLP
16
1,515.90
5.6
Bending
45.5
220
2.4




AVLLALL




AP





2
MTS
AAVLLPVL
12
1,147.40
5.6
Bending
57.3
211.7
2.3




LAAP





3
MTD
LGGAVVA
16
1,333.50
5.5
Bending
47.9
140.6
1.8



10
APVAAAV




AP





4
MTD
LAAAALA
11
1,022.30
5.5
Bending
26.6
213.6
2.4



13
VLPL





5
MTD
AAAVPVL
10
881
5.6
Bending
47.5
176
2.4



47
VAA





6
MTD
VLLAAALIA
9
854.1
5.5
No-Bending
8.9
250
3



56





7
MTD
PVLLLLA
7
737.9
6
No-Bending
36.1
278.6
2.8



73





8
MTD
AVALLILAV
9
882.1
5.6
No-Bending
30.3
271.1
3.3



77





9
MTD
AVALVAV
11
982.2
5.6
No-Bending
9.1
212.7
3.1



84
VAVA





10
MTD
LLAAAAA
11
1,010.20
5.5
No-Bending
9.1
231.8
2.7



85
LLLA





11
MTD
LALPVLLLA
11
1,010.20
5.5
No-Bending
9.1
231.8
2.7



86





12
MTD
LALPVLLLA
9
922.2
5.5
Bending
51.7
271.1
2.8



103





13
MTD
AVVVPAIV
12
1,119.40
5.6
Bending
50.3
195
2.4



132
LAAP





14
MTD
AAAPVAA
9
1,031.40
5.5
Bending
73.1
120
1.6



151
VP





15
MTD
AVIPILAVP
9
892.1
5.6
Bending
48.5
216.7
2.4



173





16
MTD
LILLLPAV
12
1,011.80
5.5
Bending
79.1
257.3
2.6



174
ALP





17
MTD
AVLLLPAAA
9
838
5.6
Bending
51.7
206.7
2.4



181

















AVE
10.8 ±
1011 ±
5.6 ±
Proline
40.1 ±
217.9 ± 43.6
2.5 ± 0.4



2.4
189.6
0.1
Presence
21.9





















TABLE 4












SEQ



Residue
A/a Composition
Secondary
ID



















#
Peptides
Structure
A
V
L
I
P
G
Structure
Cargo
Ref.
No






















1
MTM
Aliphatic
6
2
6
0
2
0
Helix
p50
1
798




Ring


2
MTS
Aliphatic
4
2
4
0
2
0
No-Helix
CRE
2
799




Ring


3
MTD10
Aliphatic
7
4
1
0
2
2
Helix
Parkin
8
800




Ring


4
MTD13
Aliphatic
5
1
4
0
1
0
No-Helix
RUNX3
3
801




Ring


5
MTD47
Aliphatic
5
3
1
0
1
0
No-Helix
CMYC
4
802




Ring


6
MTD56
Aliphatic
4
1
3
1
0
0
Helix
ES
5
803




Ring


7
MTD73
Aliphatic
1
1
4
0
1
0
Helix
ES
5
804




Ring


8
MTD77
Aliphatic
3
2
3
1
0
0
Helix
NM23
6
805




Ring


9
MTD84
Aliphatic
5
5
1
0
0
0
Helix
OCT4
4
806




Ring


10
MTD85
Aliphatic
6
0
5
0
0
0
No-Helix
RUNX3
7
807




Ring


11
MTD86
Aliphatic
6
0
5
0
0
0
No-Helix
SOX2
7
808




Ring


12
MTD103
Aliphatic
2
1
5
0
1
0
Helix
p18
8
809




Ring


13
MTD132
Aliphatic
4
4
1
1
2
0
No-Helix
LIN28
4
810




Ring


14
MTD151
Aliphatic






No-Helix
Parkin
8
811




Ring


15
MTD173
Aliphatic
2
2
1
2
2
0
Helix
KLF4
4
812




Ring


16
MTD174
Aliphatic






Helix
Parkin
8
813




Ring


17
MTD181
Aliphatic
4
1
3
0
1
0
No-Helix
SOX2
4
814




Ring









Two peptide/protein analysis programs were used (ExPasy: SoSui: http://harrier.nagahama-i-bio.ac.jp/sosui/sosui_submit.html) to determine various indexes and structural features of the peptide sequences and to design new sequence. Followings are important factors analyzed.


1-2. Characteristics of Analyzed Peptides: Length, Molecular Weight and pI Value

Average length, molecular weight and pI value of the peptides analyzed were 10.8±2.4, 1,011±189.6 and 5.6±0.1, respectively (Table 5)


Table 5 summarizes Critical Factors (CFs) of published hydrophobic Cell-Penetrating Peptides (A) which were analyzed.












TABLE 5









Length:
10.8 ± 2.4



Molecular Weight:
 1,011 ± 189.6



pl:
 5.6 ± 0.1



Bending Potential (BP):
Praline presences in




the middle and/or the end




of peptides, or No Proline.



Instability Index (II):
 40.1 ± 21.9



Residue Structure &
217.9 ± 43.6



Aliphatic Index (AI):




Hydropathy (GRAVY):
 2.5 ± 0.4



Aliphatic Ring:
Non polar hydrophobic




& aliphatic amino




acid (A, V, L, I).



Secondary Structure:
α-Helix is favored




but not required.










1-3. Characteristics of Analyzed Peptides: Bending Potential—Proline Position (PP)

Bending potential (bending or no-bending) was determined based on the fact whether proline (P) exists and/or where the amino acid(s) providing bending potential to the peptide in recombinant protein is/are located. Proline differs from the other common amino acids in that its side chain is bonded to the backbone nitrogen atom as well as the alpha-carbon atom. The resulting cyclic structure markedly influences protein architecture which is often found in the bends of folded peptide/protein chain.


Eleven out of 17 were determined as ‘Bending’ peptide which means that proline is present in the middle of sequence for peptide bending and/or located at the end of the peptide for protein bending. As indicated above, peptide sequences could penetrate the plasma membrane in a “bent” configuration. Therefore, bending or no-bending potential is considered as one of the critical factors for the improvement of current hydrophobic CPPs.


1-4. Characteristics of Analyzed Peptides: Rigidity/Flexibility—Instability Index (II)

Since one of the crucial structural features of any peptide is based on the fact whether the motif is rigid or flexible, which is an intact physicochemical characteristic of the peptide sequence, instability index (II) of the sequence was determined. The index value representing rigidity/flexibility of the peptide was extremely varied (8.9 to 79.1), but average value was 40.1±21.9 which suggested that the peptide should be somehow flexible, but not too much rigid or flexible (Tables 3 and 4).


1-5. Characteristics of Analyzed Peptides: Structural Features—Structural Feature (Aliphatic Index: AI) and Hydropathy (Grand Average of Hydropathy: GRAVY)

Alanine (V), valine (V), leucine (L) and isoleucine (I) contain aliphatic side chain and are hydrophobic—that is, they have an aversion to water and like to cluster. These amino acids having hydrophobicity and aliphatic residue enable them to pack together to form compact structure with few holes. Analyzed peptide sequence showed that all composing amino acids were hydrophobic (A, V, L and I) except glycine (G) in only one out of 17 (MTD10—Tables 3 and 4) and aliphatic (A, V, L, I, and P). Their hydropathic index (Grand Average of Hydropathy: GRAVY) and aliphatic index (AI) were 2.5±0.4 and 217.9±43.6, respectively. Their amino acid composition is also indicated in the Tables 3 and 4.


1-6. Characteristics of Analyzed Peptides: Secondary Structure (Helicity)

As explained above, the CPP sequences may be supposed to penetrate the plasma membrane directly after inserting into the membranes in a “bent” configuration with hydrophobic sequences having a-helical conformation. In addition, our analysis strongly indicated that bending potential was crucial for membrane penetration. Therefore, structural analysis of the peptides was conducted to determine whether the sequences were to form helix or not. Nine peptides were helix and eight were not (Tables 3 and 4). It seems to suggest that helix structure may not be required.


1-7. Determination of Critical Factors (CFs)

In the 11 characteristics analyzed, the following 6 are selected namely “Critical Factors” for the development of new hydrophobic CPPs—advanced MTDs: amino acid length, bending potential (proline presence and location), rigidity/flexibility (instability index: II), structural feature (aliphatic index: AI), hydropathy (GRAVY) and amino acid composition/residue structure (hydrophobic and aliphatic A/a) (Tables 3 to 5).


2. Analysis of Selected Hydrophobic CPPs to Optimize ‘Critical Factors’

Since the analyzed data of the 17 different hydrophobic CPPs (analysis A, Tables 3 to 5) previously developed during the past 2 decades showed high variation and were hard to make common- or consensus- features, analysis B (Tables 6 to 8) and C (Tables 9 to 11) were also conducted to optimize the critical factors for better design of improved CPPs-aMTDs.


Therefore, 17 hydrophobic CPPs have been grouped into two groups and analyzed the groups for their characteristics in relation to the cell permeable property. The critical factors have been optimized by comparing and contrasting the analytical data of the groups and determining the common homologous features that may be critical for the cell permeable property.


2-1. Selective Analysis (B) of Peptides Used to Biologically Active Cargo Protein for in vivo


In analysis B, eight CPPs were used with each biologically active cargo in vivo. Length was 11±3.2, but 3 out of 8 CPPs possessed little bending potential. Rigidity/Flexibility (instability index: II) was 41±15, but removing one [MTD85: rigid, with minimal II (9.1)] of the peptides increased the overall instability index to 45.6±9.3. This suggested that higher flexibility (40 or higher II) is potentially be better. All other characteristics of the 8 CPPs were similar to the analysis A, including structural feature and hydropathy (Tables 6 to 8)


Tables 6 and 7 show characteristics of published hydrophobic Cell-Penetrating Peptides (B): selected CPPs that were used to each cargo in vivo.


















TABLE 6












Rigidity/
Structural










Flexibility
Feature






Molecular

Bending
(Instability
(Aliphatic
Hydropathy


#
Peptides
Sequence
Length
Weight
pI
Potential
Index: II)
Index: AI)
(GRAVY)
























1
MTM
AAVALLPAVLLALLAP
16
1,515.9
5.6
Bending
45.5
220.0
2.4





2
MTS
AAVLLPVLLAAP
12
1,147.4
5.6
Bending
57.3
211.7
2.3





3
MTD10
LGGAVVAAPVAAAVAP
16
1,333.5
5.5
Bending
47.9
140.6
1.8





4
MTD73
PVLLLLA
7
737.9
6.0
No-
36.1
278.6
2.8








Bending





5
MTD77
AVALLILAV
9
882.1
5.6
No-
30.3
271.1
3.3








Bending





6
MTD85
LLAAAAALLLA
11
1,010.2
5.5
No-
9.1*
231.8
2.7








Bending





7
MTD103
LALPVLLLA
9
922.2
5.5
Bending
51.7
271.1
2.8





8
MTD132
AVVVPAIVLAAP
12
1,119.4
5.6
Bending
50.3
195.0
2.4

















AVE
11 ± 3.2
1,083 ± 252
5.6 ± 0.1
Proline
41 ± 15
227 ± 47
2.5 ± 0.4






Presence





*Removing the MTD85 increases II to 45.6 ± 9.3

















TABLE 7









Residue
A/a Composition
Secondary


















#
Peptides
Structure
A
V
L
I
P
G
Structure
Cargo
Ref.





1
MTM
Aliphatic
6
2
6
0
2
0
Helix
p50
1




Ring


2
MTS
Aliphatic
4
2
4
0
2
0
No-Helix
CRE
2




Ring


3
MTD10
Aliphatic
7
4
1
0
2
2
Helix
Parkin
8




Ring


4
MTD73
Aliphatic
1
1
4
0
1
0
Helix
ES
6




Ring


5
MTD77
Aliphatic
3
2
3
1
0
0
Helix
NM23
3




Ring


6
MTD85
Aliphatic
6
0
5
0
0
0
No-Helix
RUNX3
5




Ring


7
MTD103
Aliphatic
2
1
5
0
1
0
Helix
p18
4




Ring


8
MTD132
Aliphatic
4
4
1
1
2
0
No-Helix
LIN28
7




Ring









Table 8 shows summarized Critical Factors of published hydrophobic Cell-Penetrating Peptides (B).












TABLE 8









Length:
  11 ± 3.2



Molecular Weight:
1,083 ± 252



pl:
  5.6 ± 0.1



Bending Potential (BP):
Proline presences




in the middle and/or




the end of peptides,




or No Proline.



Instability Index (II):
41.0 ± 15




(a Removing the




MTD85 increases




II to 45.6 ± 9.3)



Residue Structure &
 227 ± 47



Aliphatic Index (Al):




Hydropathy (GRAVY):
  2.5 ± 0.4



Aliphatic Ring:
Non-polar hydrophobic




& aliphatic amino




acid (A, V, L, I).



Secondary Structure:
α-Helix is favored




but not required.











2-2. Selective Analysis (C) of Peptides that Provided Bending Potential and Higher Flexibility


To optimize the ‘Common Range and/or Consensus Feature of Critical Factor’ for the practical design of aMTDs and the random peptides (rPs or rPeptides), which were to prove that the ‘Critical Factors’ determined in the analysis A, B and C were correct to improve the current problems of hydrophobic CPPs—protein aggregation, low solubility/yield, and poor cell-/tissue-permeability of the recombinant proteins fused to the MTS/MTM or MTD, and non-common sequence and non-homologous structure of the peptides, empirically selected peptides were analyzed for their structural features and physicochemical factor indexes.


Hydrophobic CPPs which did not have a bending potential, rigid or too much flexible sequences (too much low or too much high Instability Index), or too low or too high hydrophobic CPPs were unselected, but secondary structure was not considered because helix structure of sequence was not required.


In analysis C, eight selected CPP sequences that could provide a bending potential and higher flexibility were finally analyzed (Tables 9 to 11). Common amino acid length is 12 (11.6±3.0). Proline is presence in the middle of and/or the end of sequence. Rigidity/Flexibility (II) is 45.5-57.3 (Avg: 50.1±3.6). AI and GRAVY representing structural feature and hydrophobicity of the peptide are 204.7±37.5 and 2.4±0.3, respectively. All peptides are consisted with hydrophobic and aliphatic amino acids (A, V, L, I, and P). Therefore, analysis C was chosen as a standard for the new design of new hydrophobic CPPs-aMTDs.


Tables 9 and 10 show characteristics of published hydrophobic Cell-Penetrating Peptides (C): selected CPPs that provided bending potential and higher flexibility.


















TABLE 9












Rigidity/











Flexibility
Structural Feature






Molecular

Bending
(Instability
(Aliphatic
Hydropathy


#
Peptides
Sequence
Length
Weight
pI
Potential
Index: II)
Index: AI)
(GRAVY)
























1
MTM
AAVALLPAVLLALLAP
16
1515.9
5.6
Bending
45.5
220.0
2.4





2
MTS
AAVLLPVLLAAP
12
1147.4
5.6
Bending
57.3
211.7
2.3





3
MTD10
LGGAVVAAPVAAAVAP
16
1333.5
5.5
Bending
47.9
140.6
1.8





4
MTD47
AAAVPVLVAA
10
881.0
5.6
Bending
47.5
176.0
2.4





5
MTD103
LALPVLLLA
9
922.2
5.5
Bending
51.7
271.1
2.8





6
MTD132
AVVVPAIVLAAP
12
1119.4
5.6
Bending
50.3
195.0
2.4





7
MTD173
AVIPILAVP
9
892.1
5.6
Bending
48.5
216.7
2.4





8
MTD181
AVLLLPAAA
9
838.0
5.6
Bending
51.7
206.7
2.4

















AVE
11.6 ±
1081.2 ±
5.6 ±
Proline
50.1 ± 3.6
204.7 ± 37.5
2.4 ± 0.3



3.0
244.6
0.1
Presence




















TABLE 10










A/a




Residue
Composition
Secondary



















#
Peptides
Sequence
Structure
A
V
L
I
P
G
Structure
Cargo
Ref.





1
MTM
AAVALLPAVLLALLAP
Aliphatic
6
2
6
0
2
0
Helix
p50
1





Ring





2
MTS
AAVLLPVLLAAP
Aliphatic
4
2
4
0
2
0
No-Helix
CRE
2





Ring





3
MTD10
LGGAVVAAPVAAAVAP
Aliphatic
7
4
1
0
2
2
Helix
Parkin
8





Ring





4
MTD47
AAAVPVLVAA
Aliphatic
5
3
1
0
1
0
No-Helix
CMYC
4





Ring





5
MTD103
LALPVLLLA
Aliphatic
2
1
5
0
1
0
Helix
p18
8





Ring





6
MTD132
AVVVPAIVLAAP
Aliphatic
4
4
1
1
2
0
No-Helix
LIN28
4





Ring





7
MTD173
AVIPILAVP
Aliphatic
2
2
1
2
2
0
Helix
KLF4
4





Ring





8
MTD181
AVLLLPAAA
Aliphatic
4
1
3
0
1
0
No-Helix
SOX2
4





Ring









Table 11 shows summarized critical factors of published hydrophobic Cell-Penetrating Peptides (C).












TABLE 11









Length:
11.6 ± 3.0



Molecular Weight:
1,081.2 ± 224.6



pl:
  5.6 ± 0.1



Bending Potential (BP):
Proline presences in




the middle and/or




the end of peptides.



Instability Index (II):
50.1 ± 3.6



Residue Structure &
204.7 ± 37.5



Aliphatic Index (AI):




Hydropathy (GRAVY):
 2.4 ± 0.3



Aliphatic Ring:
Non-polar hydrophobic




& aliphatic amino




acid (A, V, L, I).



Secondary Structure:
α-Helix is favored




but not required.










3. New Design of Improved Hydrophobic CPPs-aMTDs Based on the Optimized Critical Factors

3-1. Determination of Common Sequence and/or Common Homologous Structure


As mentioned above, H-regions of signal sequence (HRSS)-derived CPPs (MTS/MTM and MTD) do not have a common sequence, sequence motif, and/or common-structural homologous feature. In this invention, the aim is to develop improved hydrophobic CPPs formatted in the common sequence- and structural-motif which satisfy newly determined ‘Critical Factors’ to have ‘Common Function,’ namely, to facilitate protein translocation across the membrane with similar mechanism to the analyzed reference CPPs. Based on the analysis A, B and C, the common homologous features have been analyzed to determine the critical factors that influence the cell-permeability. The range value of each critical factor has been determined to include the analyzed index of each critical factor from analysis A, B and C to design novel aMTDs (Table 12). These features have been confirmed experimentally with newly designed aMTDs in their cell-permeability.


Table 12 shows comparison the range/feature of each Critical Factor between the value of analyzed CPPs and the value determined for new design of novel aMTDs sequences









TABLE 12







Summarized critical Factors of aMTD










Selected CPPs
Newly Designed CPPs


Critical Factor
Range
Range





Bending Potential
Proline presences in
Proline presences


(Proline Position: PP)
the middle and/or at
in the middle



the end of peptides
(5’, 6’, 7’ or 8’)




and at the




end of peptides


Rigidity/Flexibility
45.5-57.3
40-60


(Instability Index: II)
 (50.1 ± 3.6)



Structural Feature
140.6-220.0
180-220


(Aliphatic Index: AI)
(204.7 ± 37.5)



Hydropathy
1.8-2.8
2.1-2.6


(Grand Average of
  (2.4 ± 0.3)



Hydropathy GRAVY)




Length
 11.6 ± 3.0
 9-13


(Number of Amino Acid)




Amino acid Composition
A, V, I, L, P
A, V, I, L, P









In Table 12, universal common features and sequence/structural motif are provided. Length is 9-13 amino acids, and bending potential is provided with the presence of proline in the middle of sequence (at 5′, 6′, 7′ or 8′ amino acid) for peptide bending and at the end of peptide for recombinant protein bending and Rigidity/Flexibility of aMTDs is II>40 are described in Table 12.


3-2. Critical Factors for Development of Advanced MTDs

Recombinant cell-permeable proteins fused to the hydrophobic CPPs to deliver therapeutically active cargo molecules including proteins into live cells had previously been reported, but the fusion proteins expressed in bacteria system were hard to be purified as a soluble form due to their low solubility and yield. To address the crucial weakness for further clinical development of the cell-permeable proteins as protein-based biotherapeutics, greatly improved form of the hydrophobic CPP, named as advanced MTD (aMTD) has newly been developed through critical factors-based peptide analysis. The critical factors used for the current invention of the aMTDs are herein (Table 12).


1. Amino Acid Length: 9 to 13


2. Bending Potential (Proline Position: PP):Proline presences in the middle (from 5′ to 8′ amino acid) and at the end of sequence


3. Rigidity/Flexibility (Instability Index: II): 40 to 60


4. Structural Feature (Aliphatic Index: AI): 180 to 220


5. Hydropathy (GRAVY): 2.1 to 2.6


6. Amino Acid Composition: Hydrophobic and Aliphatic amino acids—A, V, L, I and P


3-3. Design of Potentially Best aMTDs that All Critical Factors are Considered and Satisfied


After careful consideration of six critical factors derived from analysis of unique features of hydrophobic CPPs, advanced macromolecule transduction domains (aMTDs) have been designed and developed based on the common 12 amino acid platform which satisfies the critical factors including amino acid length (9 to 13) determined from the analysis.




embedded image


Unlike previously published hydrophobic CPPs that require numerous experiments to determine their cell-permeability, newly developed aMTD sequences could be designed by performing just few steps as follows using above mentioned platform to follow the determined range value/feature of each critical factor.


First, prepare the 12 amino acid sequence platform for aMTD. Second, place proline (P) in the end (12′) of sequence and determine where to place proline in one of four U(s) in 5′, 6′, 7′, and 8. Third, alanine (A), valine (V), leucine (L) or isoleucine (I) is placed in either X(s) and/or U(s), where proline is not placed. Lastly, determine whether the amino acid sequences designed based on the platform, satisfy the value or feature of six critical factors to assure the cell permeable property of aMTD sequences. Through these processes, numerous novel aMTD sequences have been constructed. The expression vectors for preparing non-functional cargo recombinant proteins fused to each aMTD, expression vectors have been constructed and forcedly expressed in bacterial cells. These aMTD-fused recombinant proteins have been purified in soluble form and determined their cell-permeability quantitatively. aMTD sequences have been newly designed, numbered from 1 to 240, as shown in Tables 13 to 18. In Tables 13 to 18, sequence ID Number (SEQ ID NO) is a sequence listings for reference, and aMTD numbers refer to amino acid listing numbers that actually have been used at the experiments. For further experiments, aMTD numbers have been used. In addition, polynucleotide sequences shown in the sequence lists have been numbered from SEQ ID NO: 241 to 480.


Tables 13 to 18 shows 240 new hydrophobic aMTD sequences that were developed to satisfy all critical factors.
















TABLE 13









Rigidity/
Sturctural




Sequence



Flexibility
Feature
Hydropathy
Residue


ID Number
aMTD
Sequences
Length
(II)
(AI)
(GRAVY)
Structure






















1
1
AAALAPVVLALP
12
57.3
187.5
2.1
Aliphatic





2
2
AAAVPLLAVVVP
12
41.3
195.0
2.4
Aliphatic





3
3
AALLVPAAVLAP
12
57.3
187.5
2.1
Aliphatic





4
4
ALALLPVAALAP
12
57.3
195.8
2.1
Aliphatic





5
5
AAALLPVALVAP
12
57.3
187.5
2.1
Aliphatic





6
11
VVALAPALAALP
12
57.3
187.5
2.1
Aliphatic





7
12
LLAAVPAVLLAP
12
57.3
211.7
2.3
Aliphatic





8
13
AAALVPVVALLP
12
57.3
203.3
2.3
Aliphatic





9
21
AVALLPALLAVP
12
57.3
211.7
2.3
Aliphatic





10
22
AVVLVPVLAAAP
12
57.3
195.0
2.4
Aliphatic





11
23
VVLVLPAAAAVP
12
57.3
195.0
2.4
Aliphatic





12
24
IALAAPALIVAP
12
50.2
195.8
2.2
Aliphatic





13
25
IVAVAPALVALP
12
50.2
203.3
2.4
Aliphatic





14
42
VAALPVVAVVAP
12
57.3
186.7
2.4
Aliphatic





15
43
LLAAPLVVAAVP
12
41.3
187.5
2.1
Aliphatic





16
44
ALAVPVALLVAP
12
57.3
203.3
2.3
Aliphatic





17
61
VAALPVLLAALP
12
57.3
211.7
2.3
Aliphatic





18
62
VALLAPVALAVP
12
57.3
203.3
2.3
Aliphatic





19
63
AALLVPALVAVP
12
57.3
203.3
2.3
Aliphatic























TABLE 14









Rigidity/
Sturctural




Sequence



Flexibility
Feature
Hydropathy
Residue


ID Number
aMTD
Sequences
Length
(II)
(AI)
(GRAVY)
Structure







20
64
AIVALPVAVLAP
12
50.2
203.3
2.4
Aliphatic





21
65
IAIVAPVVALAP
12
50.2
203.3
2.4
Aliphatic





22
81
AALLPALAALLP
12
57.3
204.2
2.1
Aliphatic





23
82
AVVLAPVAAVLP
12
57.3
195.0
2.4
Aliphatic





24
83
LAVAAPILALALP
12
41.3
195.8
2.1
Aliphatic





25
84
AAVAAPLLLALP
12
41.3
195.8
2.1
Aliphatic





26
85
LLVLPAAALAAP
12
57.3
195.8
2.1
Aliphatic





27
101
LVALAPVAAVLP
12
57.3
203.3
2.3
Aliphatic





28
102
LALAPAALALLP
12
57.3
204.2
2.1
Aliphatic





29
103
ALIAAPILALAP
12
57.3
204.2
2.2
Aliphatic





30
104
AVVAAPLVLALP
12
41.3
203.3
2.3
Aliphatic





31
105
LLALAPAALLAP
12
57.3
204.1
2.1
Aliphatic





32
121
AIVALPALALAP
12
50.2
195.8
2.2
Aliphatic





33
123
AAIIVPAALLAP
12
50.2
195.8
2.2
Aliphatic





34
124
IAVALPALIAAP
12
50.3
195.8
2.2
Aliphatic





35
141
AVIVLPALAVAP
12
50.2
203.3
2.4
Aliphatic





36
143
AVLAVPAVLVAP
12
57.3
195.0
2.4
Aliphatic





37
144
VLAIVPAVALAP
12
50.2
203.3
2.4
Aliphatic





38
145
LLAVVPAVALAP
12
57.3
203.3
2.3
Aliphatic





39
161
AVIALPALIAAP
12
57.3
195.8
2.2
Aliphatic





40
162
AVVALPAALIVP
12
50.2
203.3
2.4
Aliphatic





41
163
LALVLPAALAAP
12
57.3
195.8
2.1
Aliphatic





42
164
LAAVLPALLAAP
12
57.3
195.8
2.1
Aliphatic





43
165
ALAVPVALAIVP
12
50.2
203.3
2.4
Aliphatic





44
182
ALIAPVVALVAP
12
57.3
203.3
2.4
Aliphatic





45
183
LLAAPVVIALAP
12
57.3
211.6
2.4
Aliphatic





46
184
LAAIVPAIIAVP
12
50.2
211.6
2.4
Aliphatic





47
185
AALVLPLIIAAP
12
41.3
220.0
2.4
Aliphatic





48
201
LALAVPALAALP
12
57.3
195.8
2.1
Aliphatic





49
204
LIAALPAVAALP
12
57.3
195.8
2.2
Aliphatic





50
205
ALALVPAIAALP
12
57.3
195.8
2.2
Aliphatic





51
221
AAILAPIVALAP
12
50.2
195.8
2.2
Aliphatic





52
222
ALLIAPAAVIAP
12
57.3
195.8
2.2
Aliphatic





53
223
AILAVPIAVVAP
12
57.3
203.3
2.4
Aliphatic





54
224
ILAAVPIALAAP
12
57.3
195.8
2.2
Aliphatic





55
225
VAALLPAAAVLP
12
57.3
187.5
2.1
Aliphatic





56
241
AAAVVPVLLVAP
12
57.3
195.0
2.4
Aliphatic





57
242
AALLVPALVAAP
12
57.3
187.5
2.1
Aliphatic





58
243
AAVLLPVALAAP
12
57.3
187.5
2.1
Aliphatic





59
245
AAALAPVLALVP
12
57.3
187.5
2.1
Aliphatic





60
261
LVLVPLLAAAAP
12
41.3
211.6
2.3
Aliphatic





61
262
ALIAVPAIIVAP
12
50.2
211.6
2.4
Aliphatic





62
263
ALAVIPAAAILP
12
54.9
195.8
2.2
Aliphatic





63
264
LAAAPVVIVIAP
12
50.2
203.3
2.4
Aliphatic





64
265
VLAIAPLLAAVP
12
41.3
211.6
2.3
Aliphatic





65
281
ALIVLPAAVAVP
12
50.2
2032
24
Aliphatic





66
282
VLAVAPALIVAP
12
50.2
203.3
2.4
Aliphatic





67
283
AALLAPALIVAP
12
50.2
195.8
2.2
Aliphatic





68
284
ALIAPAVALIVP
12
50.2
211.7
2.4
Aliphatic





69
285
AIVLLPAAVVAP
12
50.2
203.3
2.4
Aliphatic























TABLE 15









Rigidity/
Sturctural




Sequence



Flexibility
Feature
Hydropathy
Residue


ID Number
aMTD
Sequences
Length
(II)
(AI)
(GRAVY)
Structure






















70
301
VIAAPVLAVLAP
12
57.3
203.3
2.4
Aliphatic





71
302
LALAPALALLAP
12
57.3
204.2
2.1
Aliphatic





72
304
AIILAPIAAIAP
12
57.3
204.2
2.3
Aliphatic





73
305
IALAAPILLAAP
12
57.3
204.2
2.2
Aliphatic





74
321
IVAVALPALAVP
12
50.2
203.3
2.3
Aliphatic





75
322
VVAIVLPALAAP
12
50.2
203.3
2.3
Aliphatic





76
323
IVAVALPVALAP
12
50.2
203.3
2.3
Aliphatic





77
324
IVAVALPAALVP
12
50.2
203.3
2.3
Aliphatic





78
325
IVAVALPAVALP
12
50.2
203.3
2.3
Aliphatic





79
341
IVAVALPAVLAP
12
50.2
203.3
2.3
Aliphatic





80
342
VIVALAPAVLAP
12
50.2
203.3
2.3
Aliphatic





81
343
IVAVALPALVAP
12
50.2
203.3
2.3
Aliphatic





82
345
ALLIVAPVAVAP
12
50.2
203.3
2.3
Aliphatic





83
361
AVVIVAPAVIAP
12
50.2
195.0
2.4
Aliphatic





84
363
AVLAVAPALIVP
12
50.2
203.3
2.3
Aliphatic





85
364
LVAAVAPALIVP
12
50.2
203.3
2.3
Aliphatic





86
365
AVIVVAPALLAP
12
50.2
203.3
2.3
Aliphatic





87
381
VVAIVLPAVAAP
12
50.2
195.0
2.4
Aliphatic





88
382
AAALVIPAILAP
12
54.9
195.8
2.2
Aliphatic





89
383
VIVALAPALLAP
12
50.2
211.6
2.3
Aliphatic





90
384
VIVAIAPALLAP
12
50.2
211.6
2.4
Aliphatic





91
385
IVAIAVPALVAP
12
50.2
203.3
2.4
Aliphatic





92
401
AALAVIPAAILP
12
54.9
195.8
2.4
Aliphatic





93
402
ALAAVIPAAILP
12
54.9
195.8
2.2
Aliphatic





94
403
AAALVIPAAILP
12
54.9
195.8
2.2
Aliphatic





95
404
LAAAVIPAAILP
12
54.9
195.8
2.2
Aliphatic





96
405
LAAAVIPVAILP
12
54.9
211.7
2.4
Aliphatic





97
421
AAILAAPLIAVP
12
57.3
195.8
2.2
Aliphatic





98
422
VVAILAPLLAAP
12
57.3
211.7
2.4
Aliphatic





99
424
AVVVAAPVLALP
12
57.3
195.0
2.4
Aliphatic





100
425
AVVAIAPVLALP
12
57.3
203.3
2.4
Aliphatic





101
442
ALAALVPAVLVP
12
57.3
203.3
2.3
Aliphatic





102
443
ALAALVPVALVP
12
57.3
203.3
2.3
Aliphatic





103
444
LAAALVPVALVP
12
57.3
203.3
2.3
Aliphatic





104
445
ALAALVPALVVP
12
57.3
203.3
2.3
Aliphatic





105
461
IAAVIVPAVALP
12
50.2
203.3
2.4
Aliphatic





106
462
IAAVLVPAVALP
12
57.3
203.3
2.4
Aliphatic





107
463
AVAILVPLLAAP
12
57.3
211.7
2.4
Aliphatic





108
464
AVVILVPLAAAP
12
57.3
203.3
2.4
Aliphatic





109
465
IAAVIVPVAALP
12
50.2
203.3
2.4
Aliphatic





110
481
AIAIAIVPVALP
12
50.2
211.6
2.4
Aliphatic





111
482
ILAVAAIPVAVP
12
54.9
203.3
2.4
Aliphatic





112
483
ILAAAIIPAALP
12
54.9
204.1
2.2
Aliphatic





113
484
LAVVLAAPAIVP
12
50.2
203.3
2.4
Aliphatic





114
485
AILAAIVPLPVP
12
50.2
211.6
2.4
Aliphatic





115
601
VIVALAVPALAP
12
50.2
203.3
2.4
Aliphatic





116
502
AIVALAVPVLAP
12
50.2
203.3
2.4
Aliphatic





117
503
AAIIIVLPAALP
12
50.2
220.0
2.4
Aliphatic





118
604
LIVALAVPALAP
12
50.2
211.7
2.4
Aliphatic





119
505
AIIIVIAPAAAP
12
50.2
195.8
2.3
Aliphatic























TABLE 16









Rigidity/
Sturctural




Sequence



Flexibility
Feature
Hydropathy
Residue


ID Number
aMTD
Sequences
Length
(II)
(AI)
(GRAVY)
Structure







120
521
LAALIVVPAVAP
12
50.2
203.3
2.4
Aliphatic





121
522
ALLVIAVPAVAP
12
57.3
203.3
2.4
Aliphatic





122
524
AVALIVVPALAP
12
50.2
203.3
2.4
Aliphatic





123
525
ALAIVVAPVAVP
12
50.2
195.0
2.4
Aliphatic





124
541
LLALIIAPAAAP
12
57.3
204.1
2.1
Aliphatic





125
542
ALALIIVPAVAP
12
50.2
211.6
2.4
Aliphatic





126
543
LLAALIAPAALP
12
57.3
204.1
2.1
Aliphatic





127
544
IVALIVAPAAVP
12
43.1
203.3
2.4
Aliphatic





128
545
VVLVLAAPAAVP
12
57.3
195.0
2.3
Aliphatic





129
561
AAVAIVLPAVVP
12
50.2
195.0
2.4
Aliphatic





130
562
ALIAAIVPALVP
12
50.2
211.7
2.4
Aliphatic





131
563
ALAVIVVPALAP
12
50.2
203.3
2.4
Aliphatic





132
564
VAIALIVPALAP
12
50.2
211.7
2.4
Aliphatic





133
565
VAIVLVAPAVAP
12
50.2
195.0
2.4
Aliphatic





134
582
VAVALIVPALAP
12
50.2
203.3
2.4
Aliphatic





135
583
AVILALAPIVAP
12
50.2
211.6
2.4
Aliphatic





136
585
ALIVAIAPALVP
12
50.2
211.6
2.4
Aliphatic





137
601
AAILIAVPIAAP
12
57.3
195.8
2.3
Aliphatic





138
602
VIVALAAPVLAP
12
50.2
203.3
2.4
Aliphatic





139
603
VLVALAAPVIAP
12
57.3
203.3
2.4
Aliphatic





140
604
VALIAVAPAVVP
12
57.3
195.0
2.4
Aliphatic





141
605
VIAAVLAPVAVP
12
57.3
195.0
2.4
Aliphatic





142
622
ALIVLAAPVAVP
12
50.2
203.3
2.4
Aliphatic





143
623
VAAAIALPAIVP
12
50.2
187.5
2.3
Aliphatic





144
625
ILAAAAAPLIVP
12
50.2
195.8
2.2
Aliphatic





145
643
LALVLAAPAIVP
12
50.2
211.6
2.4
Aliphatic





146
645
ALAVVALPAIVP
12
50.2
203.3
2.4
Aliphatic





147
661
AAILAPIVAALP
12
50.2
195.8
2.2
Aliphatic





148
664
ILIAIAIPAAAP
12
54.9
204.1
2.3
Aliphatic





149
665
LAIVLAAPVAVP
12
50.2
203.3
2.3
Aliphatic





150
666
AAIAIIAPAIVP
12
50.2
195.8
2.3
Aliphatic





151
667
LAVAIVAPALVP
12
50.2
203.3
2.3
Aliphatic





152
683
LAIVLAAPAVLP
12
50.2
211.7
2.4
Aliphatic





153
684
AAIVLALPAVLP
12
50.2
211.7
2.4
Aliphatic





154
685
ALLVAVLPAALP
12
57.3
211.7
2.3
Aliphatic





155
686
AALVAVLPVALP
12
57.3
203.3
2.3
Aliphatic





156
687
AILAVALPLLAP
12
57.3
220.0
2.3
Aliphatic





157
703
IVAVALVPALAP
12
50.2
203.3
2.4
Aliphatic





158
705
IVAVALLPALAP
12
50.2
211.7
2.4
Aliphatic





159
706
IVAVALLPAVAP
12
50.2
203.3
2.4
Aliphatic





160
707
IVALAVLPAVAP
12
50.2
203.3
2.4
Aliphatic





161
724
VAVLAVLPALAP
12
57.3
203.3
2.3
Aliphatic





162
725
IAVLAVAPAVLP
12
57.3
203.3
2.3
Aliphatic





163
726
LAVAIIAPAVAP
12
57.3
187.5
2.2
Aliphatic





164
727
VALAIALPAVLP
12
57.3
211.6
2.3
Aliphatic





165
743
AIAIALVPVALP
12
57.3
211.6
2.4
Aliphatic





166
744
AAVVIVAPVALP
12
50.2
195.0
2.4
Aliphatic





167
746
VAIIVVAPALAP
12
50.2
203.3
2.4
Aliphatic





168
747
VALLAIAPALAP
12
57.3
195.8
2.2
Aliphatic





169
763
VAVLIAVPALAP
12
57.3
203.3
2.3
Aliphatic























TABLE 17









Rigidity/
Sturctural




Sequence



Flexibility
Feature
Hydropathy
Residue


ID Number
aMTD
Sequences
Length
(II)
(AI)
(GRAVY)
Structure







170
764
AVALAVLPAVVP
12
57.3
195.0
2.3
Aliphatic





171
765
AVALAVVPAVLP
12
57.3
195.0
2.3
Aliphatic





172
766
IVVIAVAPAVAP
12
50.2
195.0
2.4
Aliphatic





173
767
IVVAAVVPALAP
12
50.2
195.0
2.4
Aliphatic





174
783
IVALVPAVAIAP
12
50.2
203.3
2.5
Aliphatic





175
784
VAALPAVALVVP
12
57.3
195.0
2.4
Aliphatic





176
786
LVAIAPLAVLAP
12
41.3
211.7
2.4
Aliphatic





177
787
AVALVPVIVAAP
12
502
195.0
2.4
Aliphatic





178
788
AIAVAIAPVALP
12
57.3
187.5
2.3
Aliphatic





179
803
AIALAVPVLALP
12
57.3
211.7
2.4
Aliphatic





180
805
LVLIAAAPIALP
12
41.3
220.0
2.4
Aliphatic





181
806
LVALAVPAAVLP
12
57.3
203.3
2.3
Aliphatic





182
807
AVALAVPALVLP
12
57.3
203.3
2.3
Aliphatic





183
808
LVVLAAAPLAVP
12
41.3
203.3
2.3
Aliphatic





184
809
LIVLAAPALAAP
12
50.2
195.8
2.2
Aliphatic





185
810
VIVLAAPALAAP
12
50.2
187.5
2.2
Aliphatic





186
811
AVVLAVPALAVP
12
57.3
195.0
2.3
Aliphatic





187
824
LIIVAAAPAVAP
12
50.2
187.5
2.3
Aliphatic





188
825
IVAVIVAPAVAP
12
43.2
195.0
2.5
Aliphatic





189
826
LVALAAPIIAVP
12
41.3
211.7
2.4
Aliphatic





190
827
IAAVLAAPALVP
12
57.3
187.5
2.2
Aliphatic





191
828
IALLAAPIIAVP
12
41.3
220.0
2.4
Aliphatic





192
829
AALALVAPVIVP
12
50.2
203.3
2.4
Aliphatic





193
830
IALVAAPVALVP
12
57.3
203.3
2.4
Aliphatic





194
831
IIVAVAPAAIVP
12
43.2
203.3
2.5
Aliphatic





195
832
AVAAIVPVIVAP
12
43.2
195.0
2.5
Aliphatic





196
843
AVLVLVAPAAAP
12
41.3
219.2
2.5
Aliphatic





197
844
VVALLAPLIAAP
12
41.3
211.8
2.4
Aliphatic





198
845
AAVVIAPLLAVP
12
41.2
203.3
2.4
Aliphatic





199
846
IAVAVAAPLLVP
12
41.3
203.3
2.4
Aliphatic





200
847
LVAIVVLPAVAP
12
50.2
219.2
2.6
Aliphatic





201
848
AVAIVVLPAVAP
12
50.2
195.0
2.4
Aliphatic





202
849
AVILLAPLIAAP
12
57.3
220.0
2.4
Aliphatic





203
850
LVIALAAVALP
12
57.3
211.7
2.4
Aliphatic





204
851
VLAVVLPAVALP
12
57.3
219.2
2.5
Aliphatic





205
852
VLAVAAPAVLLP
12
57.3
203.3
2.3
Aliphatic





206
863
AAVVLLPIIAAP
12
41.3
211.7
2.4
Aliphatic





207
864
ALLVIAPAIAVP
12
57.3
211.7
2.4
Aliphatic





208
865
AVLVIAVPAIAP
12
57.3
203.3
2.5
Aliphatic





209
867
ALLVVIAPLAAP
12
41.3
211.7
2.4
Aliphatic





210
868
VLVAAILPAAIP
12
54.9
211.7
2.4
Aliphatic





211
870
VLVAAVLPIAAP
12
41.3
203.3
2.4
Aliphatic





212
872
VLAAAVLPLVVP
12
41.3
219.2
2.5
Aliphatic





213
875
AIAIVVPAVAVP
12
50.2
195.0
2.4
Aliphatic





214
877
VAIIAVPAVVAP
12
572
195.0
2.4
Aliphatic





215
878
IVALVAPAAVVP
12
50.2
195.0
2.4
Aliphatic





216
879
AAIVILLPAVVVP
12
50.2
219.1
2.5
Aliphatic





217
881
AALIVVPAVAVP
12
50.2
1950
2.4
Aliphatic





218
882
AIALVVPAVAVP
12
57.3
195.0
2.4
Aliphatic





219
883
LAIVPAAIAALP
12
50.2
195.8
2.2
Aliphatic























TABLE 18









Rigidity/
Sturctural




Sequence



Flexibility
Feature
Hydropathy
Residue


ID Number
aMTD
Sequences
Length
(II)
(AI)
(GRAVY)
Structure







220
885
LVAIAPAVAVLP
12
57.3
203.3
2.4
Aliphatic





221
887
VLAVAPAVAVLP
12
57.3
195.0
2.4
Aliphatic





222
888
ILAVVAIPAAAP
12
54.9
187.5
2.3
Aliphatic





223
889
ILVAAAPIAALP
12
57.3
195.8
2.2
Aliphatic





224
891
ILAVAAIPAALP
12
54.9
195.8
2.2
Aliphatic





225
893
VIAIPAILAAAP
12
54.9
195.8
2.3
Aliphatic





226
895
AIIIVVPAIAAP
12
50.2
211.7
2.5
Aliphatic





227
896
AILIVVAPIAAP
12
50.2
211.7
2.5
Aliphatic





228
897
AVIVPVAIIAAP
12
50.2
203.3
2.5
Aliphatic





229
899
AVVIALPAVVAP
12
57.3
195.0
2.4
Aliphatic





230
900
ALVAVIAPVVAP
12
57.3
195.0
2.4
Aliphatic





231
901
ALVAVLPAVAVP
12
57.3
195.0
2.4
Aliphatic





232
902
ALVAPLLAVAVP
12
41.3
203.3
2.3
Aliphatic





233
904
AVLAVVAPVVAP
12
57.3
186.7
2.4
Aliphatic





234
905
AVIAVAPLVVAP
12
41.3
195.0
2.4
Aliphatic





235
906
AVIALAPVVVAP
12
57.3
195.0
2.4
Aliphatic





236
907
VAIALAPVVVAP
12
57.3
195.0
2.4
Aliphatic





237
908
VALALAPVVVAP
12
57.3
195.0
2.3
Aliphatic





238
910
VAALLPAVVVAP
12
57.3
195.0
2.3
Aliphatic





239
911
VALALPAVVVAP
12
57.3
195.0
2.3
Aliphatic





240
912
VALLAPAVVVAP
12
57.3
195.0
2.3
Aliphatic









52.6 ± 5.1
201.7 ± 7.8
2.3 ± 0.1










3-4. Design of the Peptides that did not Satisfy at Least One Critical Factor


To demonstrate that this invention of new hydrophobic CPPs-aMTDs, which satisfy all critical factors described above, are correct and rationally designed, the peptides which do not satisfy at least one critical factor have also been designed. Total of 31 rPeptides (rPs) are designed, developed and categorized as follows: no bending peptides, either no proline in the middle as well at the end and/or no central proline; rigid peptides (II<40); too much flexible peptides; aromatic peptides (aromatic ring presences); hydrophobic, with non-aromatic peptides but have amino acids other than A, V, L, I, P or additional proline residues; hydrophilic, but non-aliphatic peptides.


3-4-1. Peptides that do not Satisfy the Bending Potential


Table 19 shows the peptides that do not have any proline in the middle (at 5′, 6′, 7′ or 8′) and at the end of the sequences (SEQ ID NOs: 815 to 824). In addition, Table 19 describes the peptides that do not have proline in the middle of the sequences. All these peptides are supposed to have no-bending potential.

















TABLE 19






SEQ



Proline
Rigidity/
Sturctural




ID
rPeptide


Position
Flexibility
Feature
Hydropathy


Group
NO
ID
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)























No-Bending
815
931
AVLIAPAILAAA
12
6
57.3
204.2
2.5


Peptides
816
936
ALLILAAAVAAP
12 
12
41.3
204.2
2.4


(No Proline
817
152
LAAAVAAVAALL
12
None
9.2
204.2
2.7


at 5 or 6
818
27
LAIVAAAAALVA
12
None
2.1
204.2
2.8


and/or 12)
819
935
ALLILPAAAVAA
12
6
57.3
204.2
2.4


and
820
670
ALLILAAAVAAL
12
None
25.2
236.6
2.8


(No Central
821
934
LILAPAAVVAAA
12
5
57.3
195.8
2.5


Proline)
822
37
TTCSQQQYCTNG
12
None
53.1
0
−1.1



823
16
NNSCTTYTNGSQ
12
None
47.4
0
−1.4



824
113
PVAVALLIAVPP
12
1, 11, 12
57.3
195
2.1










3-4-2. Peptides that do not Satisfy the Rigidity/Flexibility


To prove that rigidity/flexibility of the sequence is a crucial critical factor, rigid (Avg. II: 21.8±6.6) and too high flexible sequences (Avg. II: 82.3±21.0) were also designed. Rigid peptides that instability index is much lower than that of new aMTDs (II: 41.3 to 57.3, Avg. II: 53.3±5.7) are shown in Table 20 (SEQ ID NOs: 825 to 839). Bending, but too high flexible peptides that II is much higher than that of new aMTDs are also provided in Table 21 (SEQ ID NOs: 840 to 858).

















TABLE 20






SEQ



Proline
Rigidity/
Sturctural




ID
rPeptide


Position
Flexibility
Feature
Hydropathy


Group
NO
ID
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)























Rigid
825
226
ALVAAIPALAIP
12
6
20.4
195.8
2.2


Peptides
826
6
VIAMIPAAFWVA
12
6
15.7
146.7
2.2


(II <50)
827
750
LAIAAIAPLAIP
12
8, 12
22.8
204.2
2.2



828
26
AAIALAAPLAIV
12
8
18.1
204.2
2.5



829
527
LVLAAVAPIAIP
12
8, 12
22.8
211.7
2.4



830
466
IIAAAAPLAIIP
12
7, 12
22.8
204.2
2.3



831
167
VAIAIPAALAIP
12
6, 12
20.4
195.8
2.3



832
246
VVAVPLLVAFAA
12
5
25.2
195
2.7



833
426
AAALAIPLAIIP
12
7, 12
4.37
204.2
2.2



834
606
AAAIAAIPIIIP
12
8, 12
4.4
204.2
2.4



835
66
AGVLGGPIMGVP
12
7, 12
35.5
121.7
1.3



836
248
VAAIVPIAALVP
12
6, 12
34.2
203.3
2.5



837
227
LAAIVPIAAAVP
12
6, 12
34.2
187.5
2.2



838
17
GGCSAPQTTCSN
12
6
51.6
8.3
−0.5



839
67
LDAEVPLADDVP
12
6, 12
34.2
130
0.3
























TABLE 21






SEQ



Proline
Rigidity/
Sturctural




ID
rPeptide


Position
Flexibility
Feature
Hydropathy


Group
NO
ID
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)























Bending
840
692
PAPLPPVVILAV
12
1, 3, 5, 6
105.5
186.7
1.8


Peptides,
841
69
PVAVLPPAALVP
12
1, 6, 7, 12
89.4
162.5
1.6


but Too
842
390
VPLLVPVVPVVP
12
2, 6, 9, 12
105.4
210
2.2


High
843
350
VPILVPVVPVVP
12
2, 6, 9, 12
121.5
210
2.2


Flexibility
844
331
VPVLVPLVPVVP
12
2, 6, 9, 12
105.4
210
2.2



845
9
VALVPAALILPP
12
5, 11, 12
89.4
203.3
2.1



846
68
VAPVLPAAPLVP
12
3, 6, 9, 12
105.5
162.5
1.6



847
349
VPVLVPVVPVVP
12
2, 6, 9, 12
121.5
201.6
2.2



848
937
VPVLVPLPVPVV
12
2, 6, 8, 10
121.5
210
2.2



849
938
VPVLLPVVVPVP
12
2, 6, 10, 12
121.5
210
2.2



850
329
LPVLVPVVPVVP
12
2, 6, 9, 12
121.5
210
2.2



851
49
VVPAAPAVPVVP
12
3, 6, 9, 12
121.5
145.8
1.7



852
772
LPVAPVIPIIVP
12
2, 5, 8, 12
79.9
210.8
2.1



853
210
ALIALPALPALP
12
6, 9, 12
89.4
195.8
1.8



854
28
AVPLLPLVPAVP
12
3, 6, 9, 12
89.4
186.8
1.8



855
693
AAPVLPVAVPIV
12
3, 6, 10
82.3
186.7
2.1



856
169
VALVAPALILAP
12
6, 12
73.4
211.7
2.4



857
29
VLPPLPVLPVLP
12
3, 4, 6, 9, 12
121.5
202.5
1.7



858
190
AAILAPAVIAPP
12
6, 11, 12
89.4
163.3
1.8










3-4-3. Peptides that do not Satisfy the Structural Features


New hydrophobic CPPs-aMTDs are consisted with only hydrophobic and aliphatic amino acids (A, V, L, I and P) with average ranges of the indexes—AI: 180 to 220 and GRAVY: 2.1 to 2.6 (Table 12). Based on the structural indexes, the peptides which contain an aromatic residue (W, F or Y) are shown in Table 22 (SEQ ID NOs: 859 to 864) and the peptides which are hydrophobic with non-aromatic sequences but have amino acids residue other than A, V, L, I, P or additional proline residues are designed (Table 23) (SEQ ID NOs: 865 to 872). Finally, hydrophilic and/or bending peptides which are consisted with non-aliphatic amino acids are shown in Table 24 (SEQ ID NOs: 873 to 885).

















TABLE 22






SEQ



Proline
Rigidity/
Sturctural




ID
rPeptide


Position
Flexibility
Feature
Hydropathy


Group
NO
ID
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)























Aromatic
859
30
AMALLPAAVAVA
12
6
51.6
163.3
2.3


Peptides
860
33
AAAILAPAFLAV
12
7
57.3
171.7
2.4


(Aromatic
861
131
WIIAPVWLAWIA
12
5
51.6
179.2
1.9


Ring
862
922
WYVIFVLPLVVP
12
8, 12
41.3
194.2
2.2


Presences)
863
71
FMWMWFPFMWYP
12
7, 12
71.3
0
0.6



864
921
IWWFVVLPLVVP
12
8, 12
41.3
194.2
2.2
























TABLE 23






SEQ



Proline
Rigidity/
Sturctural




ID
rPeptide


Position
Flexibility
Feature
Hydropathy


Group
NO
ID
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)























Hydrophobic,
865
436
VVMLVVPAVMLP
12
 7, 12
57.3
194.2
2.6


but Non
866
138
PPAALLAILAVA
12
1, 2
57.3
195.8
2.2


Aromatic
867
77
PVALVLVALVAP
12
 1, 12
41.3
219.2
2.5


Peptides
868
577
MLMIALVPMIAV
12
8
18.9
195
2.7



869
97
ALLAAPPALLAL
12
6, 7
57.3
204.2
2.1



870
214
ALIVAPALMALP
12
 6, 12
60.5
187.5
2.2



871
59
AVLAAPVVAALA
12
6
41.3
187.5
2.5



872
54
LAVAAPPVVALL
12
6, 7
57.3
203.3
2.3
























TABLE 24






SEQ



Proline
Rigidity/
Sturctural




ID
rPeptide


Position
Flexibility
Feature
Hydropathy


Group
NO
ID
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)























Hydrophilic
873
949
SGNSCQQCGNSS
12
None
41.7
0
−1.1


Peptides,
874
39
CYNTSPCTGCCY
12
6
52.5
0
0


but Non
875
19
YVSCCTYTNGSQ
12
None
47.7
0
−1


Aliphatic
876
947
CYYNQQSNNNNQ
12
None
59.6
0
−2.4



877
139
TGSTNSPTCTST
12
7
53.4
0
−0.7



878
18
NYCCTPTTNGQS
12
6
47.9
0
−0.9



879
20
NYCNTCPTYGQS
12
7
47.4
0
−0.9



880
635
GSTGGSQQNNQY
12
None
31.9
0
−1.9



881
40
TYNTSCTPGTCY
12
8
49.4
0.0
−0.6



882
57
QNNCNTSSQGGG
12
None
52.4
0
−1.6



883
159
CYSGSTSQNQPP
12
11, 12
51
0
−1.3



884
700
GTSNTCQSNQNS
12
None
19.1
0
−1.6



885
38
YYNQSTCGGQCY
12
None
53.8
0
−1









3-5. Summary of Newly Designed Peptides

Total of 457 sequences have been designed based on the critical factors. Designed potentially best aMTDs (hydrophobic, flexible, bending, aliphatic and 12-A/a length peptides) that do satisfy all range/feature of critical factors are 316. Designed rPeptides that do not satisfy at least one of the critical factors are 141 that no bending peptide sequences are 26; rigid peptide (II<40) sequences are 23; too much flexible peptides are 24; aromatic peptides (aromatic ring presences) are 27; hydrophobic, but non-aromatic peptides are 23; and hydrophilic, but non-aliphatic peptides are 18.


4. Preparation of Recombinant Report Proteins Fused to aMTDs and rPeptides

Recombinant proteins fused to aMTDs and others [rPeptides, reference hydrophobic CPP sequences (MTM and MTD)] were expressed in a bacterial system, purified with single-step affinity chromatography and prepared as soluble proteins in physiological condition. These recombinant proteins have been tested for the ability of their cell-permeability by utilizing flow cytometry and laser scanning confocal microscopy.


4-1. Selection of Cargo Protein for Recombinant Proteins Fused to Peptide Sequences

For clinical/non-clinical application, aMTD-fused cargo materials would be biologically active molecules that could be one of the following: enzymes, transcription factors, toxic, antigenic peptides, antibodies and antibody fragments. Furthermore, biologically active molecules could be one of these following macromolecules: enzymes, hormones, carriers, immunoglobulin, membrane-bound proteins, transmembrane proteins, internal proteins, external proteins, secreted proteins, virus proteins, native proteins, glycoproteins, fragmented proteins, disulfide bonded proteins, recombinant proteins, chemically modified proteins and prions. In addition, these biologically active molecules could be one of the following: nucleic acid, coding nucleic acid sequence, mRNAs, antisense RNA molecule, carbohydrate, lipid and glycolipid.


According to these pre-required conditions, a non-functional cargo to evaluate aMTD-mediated protein uptake has been selected and called as Cargo A (CRA) that should be soluble and non-functional. The domain (A/a 289 to 840; 184 A/a length) is derived from protein S (Genbank ID: CP000113.1).


4-2. Construction of Expression Vector and Preparation of Recombinant Proteins

Coding sequences for recombinant proteins fused to each aMTD are cloned Ndel (5′) and SalI (3′) in pET-28a(+) (Novagen, Darmstadt, Germany) from PCR-amplified DNA segments. PCR primers for the recombinant proteins fused to aMTD and rPeptides are SEQ ID NOs: 481 to 797 and 886 to 1202. Structure of the recombinant proteins is displayed in FIG. 1.


The recombinant proteins were forcedly expressed in E. coli BL21 (DE3) cells grown to an OD600 of 0.6 and induced for 2 hours with 0.7 mM isopropyl-β-D-thiogalactopyranoside (IPTG). The proteins were purified by Ni2+ affinity chromatography as directed by the supplier (Qiagen, Hilden, Germany) in natural condition. After the purification, purified proteins were dissolved in a physiological buffer such as DMEM medium.












TABLE 25









Potentially Best aMTDs
240



(Hydrophobic, Flexible, Bending,




Aliphatic & Helical):




Random Peptides:
31



No Bending Peptides (No
02



Proline at 5 or 6 and/or 12):




No Bending Peptides
01



(No Central Proline):




Rigid Peptides (II < 50):
09



Too Much Flexible Peptides:
09



Aromatic Peptides (Aromatic
01



Ring Presences):




Hydrophobic, But Non-Aromatic
02



Peptides:




Hydrophilic, But Non-Aliphatic
07



Peptides:











4-3. Expression of aMTD- or Random Peptide (rP)-Fused Recombinant Proteins


Using the standardized six critical factors, 316 aMTD sequences have been designed. In addition, 141 rPeptides are also developed that lack one of these critical factors: no bending peptides: i) absence of proline both in the middle and at the end of sequence or ii) absence of proline either in the middle or at the end of sequence, rigid peptides, too much flexible peptides, aromatic peptides (aromatic ring presence), hydrophobic but non-aromatic peptides, and hydrophilic but non-aliphatic peptides (Table 25).


These rPeptides are devised to be compared and contrasted with aMTDs in order to analyze structure/sequence activity relationship (SAR) of each critical factor with regard to the peptides' intracellular delivery potential. All peptide (aMTD or rPeptide)-containing recombinant proteins have been fused to the CRA to enhance the solubility of the recombinant proteins to be expressed, purified, prepared and analyzed.


These designed 316 aMTDs and 141 rPeptides fused to CRA were all cloned (FIG. 2) and tested for inducible expression in E. coli (FIG. 3). Out of these peptides, 240 aMTDs were inducibly expressed, purified and prepared in soluble form (FIG. 4). In addition, 31 rPeptides were also prepared as soluble form (FIG. 4).


To prepare the proteins fused to rPeptides, 60 proteins were expressed that were 10 out of 26 rPeptides in the category of no bending peptides (Table 19); 15 out of 23 in the category of rigid peptides [instability index (II)<40] (Table 20); 19 out of 24 in the category of too much flexible peptides (Table 21); 6 out of 27 in the category of aromatic peptides (Table 22); 8 out of 23 in the category of hydrophobic but non-aromatic peptides (Table 23); and 12 out of 18 in the category of hydrophilic but non-aliphatic peptides (Table 24).


4-4. Quantitative Cell-Permeability of aMTD-Fused Recombinant Proteins


The aMTDs and rPeptides were fluorescently labeled and compared based on the critical factors for cell-permeability by using flow cytometry and confocal laser scanning microscopy (FIGS. 5 to 8). The cellular uptake of the peptide-fused non-functional cargo recombinant proteins could quantitatively be evaluated in flow cytometry, while confocal laser scanning microscopy allows intracellular uptake to be assessed visually. The analysis included recombinant proteins fused to a negative control [rP38] that has opposite characteristics (hydrophilic and aromatic sequence: YYNQSTCGGQCY) to the aMTDs (hydrophobic and aliphatic sequences). Relative cell-permeability (relative fold) of aMTDs to the negative control was also analyzed (Table 26 and FIG. 9).


Table 26 shows comparison analysis of cell-permeability of aMTDs with a negative control (A: rP38).












TABLE 26








Negative Control




rP38









aMTD
19.6 ± 1.6*



The Average of
(Best: 164.2)



240 aMTDs







*Relative Fold (aMTD in Geo Mean in its comparison to rP38)






Relative cell-permeability (relative fold) of aMTDs to the reference CPPs [B: MTM12 (AAVLLPVLLAAP), C: MTD85 (AVALLILAV)] was also analyzed.


Table 27 shows comparison analysis of cell-permeability of aMTDs with a reference CPP (B: MTM12).












TABLE 27








MTM12









aMTD
13.1 ± 1.1*



The Average of
(Best: 109.9)



240 aMTDs







*Relative Fold (aMTD in Geo Mean in its comparison to MTM12)






Table 28 shows comparison analysis of cell-permeability of aMTDs with a reference CPP (C: MTD85).












TABLE 28








MTD85









aMTD
6.6 ± 0.5*



The Average of
(Best: 55.5)



240 aMTDs







*Relative Fold (aMTD in Geo Mean in its comparison to MTD85)






Geometric means of negative control (histidine-tagged rP38-fused CRA recombinant protein) subtracted by that of naked protein (histidine-tagged CRA protein) lacking any peptide (rP38 or aMTD) was standardized as relative fold of 1. Relative cell-permeability of 240 aMTDs to the negative control (A type) was significantly increased by up to 164 fold, with average increase of 19.6±1.6 (Table 29 to 34).















TABLE 29







SEQ

Proline
Rigidity/
Sturctural

Relative


ID

Position
Flexibility
Feature
Hydropathy
Ratio (Fold)

















NO
aMTD
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)
A
B
C




















229
899
AVVIALPAVVAP
12
7
57.3
195.0
2.4
164.2
109.9
55.5





237
908
VALALAPVVVAP
12
7
57.3
195.0
2.3
150.6
100.8
50.9





238
910
VAALLPAVVVAP
12
6
57.3
195.0
2.3
148.5
99.4
50.2





185
810
VIVLAAPALAAP
12
7
50.2
187.5
2.2
120.0
80.3
40.6





233
904
AVLAVVAPVVAP
12
8
57.3
186.7
2.4
105.7
70.8
35.8





74
321
IVAVALPALAVP
12
7
50.2
203.3
2.3
97.8
65.2
32.9





204
851
VLAVVLPAVALP
12
7
57.3
219.2
2.5
96.6
64.7
32.7





239
911
VALALPAVVVAP
12
6
57.3
195.0
2.3
84.8
56.8
28.7





205
852
VLAVAAPAVLLP
12
7
57.3
203.3
2.3
84.6
56.6
28.6





179
803
AIALAVPVLALP
12
7
57.3
211.7
2.4
74.7
50.0
25.3





222
888
ILAVVAIPAAAP
12
8
54.9
187.5
2.3
71.0
47.5
24.0





188
825
IVAVIVAPAVAP
12
8
43.2
195.0
2.5
69.7
46.6
23.6





226
895
AIIIVVPAIAAP
12
7
50.2
211.7
2.5
60.8
40.7
20.6





227
896
AILIVVAPIAAP
12
8
50.2
211.7
2.5
57.5
38.5
19.4





164
727
VALAIALPAVLP
12
8
57.3
211.6
2.3
54.7
36.7
18.5





139
603
VLVALAAPVIAP
12
8
57.3
203.3
2.4
54.1
36.1
18.2





200
847
LVAIVVLPAVAP
12
8
50.2
219.2
2.6
50.2
33.4
16.9





189
826
LVALAAPIIAVP
12
7
41.3
211.7
2.4
49.2
32.9
16.6





161
724
VAVLAVLPALAP
12
8
57.3
203.3
2.3
47.5
31.8
16.1





131
563
ALAVIVVPALAP
12
8
50.2
203.3
2.4
47.1
31.4
15.9





186
811
AVVLAVPALAVP
12
7
57.3
195.0
2.3
46.5
31.1
15.7





194
831
IIVAVAPAAIVP
12
7
43.2
203.3
2.5
46.3
31.0
15.7





192
829
AALALVAPVIVP
12
8
50.2
203.3
2.4
44.8
30.0
15.2





224
891
ILAVAAIPAALP
12
8
54.9
195.8
2.2
44.7
29.9
15.1





234
905
AVIAVAPLVVAP
12
7
41.3
195.0
2.4
44.0
29.5
14.9





132
564
VAIALIVPALAP
12
8
50.2
211.7
2.4
43.6
29.1
14.7





34
124
IAVALPALIAAP
12
6
50.3
195.8
2.2
43.6
29.0
14.7





190
827
IAAVLAAPALVP
12
8
57.3
187.5
2.2
43.0
28.8
14.6





2
2
AAAVPLLAVVVP
12
5
41.3
195.0
2.4
40.9
27.2
13.8





91
385
IVAIAVPALVAP
12
7
50.2
203.3
2.4
38.8
25.9
13.1





191
828
IALLAAPIIAVP
12
7
41.3
220.0
2.4
36.8
24.6
12.4





181
806
LVALAVPAAVLP
12
7
57.3
203.3
2.3
36.7
24.6
12.4





198
845
AAVVIAPLLAVP
12
7
41.3
203.3
2.4
35.8
24.0
12.1





218
882
AIALVVPAVAVP
12
7
57.3
195.0
2.4
35.0
23.4
11.8





128
545
VVLVLAAPAAVP
12
8
57.3
195.0
2.3
34.6
23.1
11.7





39
161
AVIALPALIAAP
12
6
57.3
195.8
2.2
34.5
23.0
11.6





110
481
AIAIAIVPVALP
12
8
50.2
211.6
2.4
34.3
23.0
11.6





230
900
ALVAVIAPVVAP
12
8
57.3
195.0
2.4
34.3
22.9
11.6





53
223
AILAVPIAVVAP
12
6
57.3
203.3
2.4
33.0
22.1
11.2





187
824
LIIVAAAPAVAP
12
8
50.2
187.5
2.3
32.8
21.9
11.1





130
562
ALIAAIVPALVP
12
8
50.2
211.7
2.4
32.7
21.8
11.0





52
222
ALLIAPAAVIAP
12
6
57.3
195.8
2.2
32.6
21.7
11.0





17
61
VAALPVLLAALP
12
5
57.3
211.7
2.3
31.2
20.8
10.5





134
582
VAVALIVPALAP
12
8
50.2
203.3
2.4
30.6
20.4
10.3





223
889
ILVAAAPIAALP
12
7
57.3
195.8
2.2
30.3
20.3
10.3





177
787
AVALVPVIVAAP
12
6
50.2
195.0
2.4
29.3
19.6
9.9





157
703
IVAVALVPALAP
12
8
50.2
203.3
2.4
29.2
19.5
9.9





158
705
IVAVALLPALAP
12
8
50.2
211.7
2.4
28.6
19.1
9.7





220
885
LVAIAPAVAVLP
12
6
57.3
203.3
2.4
28.3
19.0
9.6





3
3
AALLVPAAVLAP
12
6
57.3
187.5
2.1
27.0
18.0
9.1





137
601
AAILIAVPIAAP
12
8
57.3
195.8
2.3
26.8
17.9
9.0





196
843
AVLVLVAPAAAP
12
8
41.3
219.2
2.5
26.4
17.7
8.9





94
403
AAALVIPAAILP
12
7
54.9
195.8
2.2
25.2
16.8
8.5





127
544
IVALIVAPAAVP
12
8
43.1
203.3
2.4
23.4
15.6
7.9





121
522
ALLVIAVPAVAP
12
8
57.3
203.3
2.4
22.7
15.2
7.7






















TABLE 30













Relative


SEQ

Proline
Rigidity/
Sturctural

Ratio


ID

Position
Flexibility
Feature
Hydropathy
(Fold)

















NO
aMTD
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)
A
B
C




















180
805
LVLIAAAPIALP
12
8
41.3
220.0
2.4
22.3
14.9
7.6





108
464
AVVILVPLAAAP
12
7
57.3
203.3
2.4
22.3
14.9
7.5





96
405
LAAAVIPVAILP
12
7
54.9
211.7
2.4
22.2
14.8
7.5





168
747
VALLAIAPALAP
12
8
57.3
195.8
2.2
22.0
14.8
7.5





115
501
VIVALAVPALAP
12
8
50.2
203.3
2.4
21.5
14.4
7.3





147
661
AAILAPIVAALP
12
6
50.2
195.8
2.2
21.4
14.3
7.2





176
786
LVAIAPLAVLAP
12
6
41.3
211.7
2.4
21.2
14.2
7.2





144
625
ILAAAAAPLIVP
12
8
50.2
195.8
2.2
20.9
13.9
7.0





101
442
ALAALVPAVLVP
12
7
57.3
203.3
2.3
20.4
13.6
6.9





240
912
VALLAPAVVVAP
12
6
57.3
195.0
2.3
19.9
13.3
6.7





43
165
ALAVPVALAIVP
12
5
50.2
203.3
2.4
19.8
13.2
6.7





98
422
VVAILAPLLAAP
12
7
57.3
211.7
2.4
19.6
13.1
6.6





155
686
AALVAVLPVALP
12
8
57.3
203.3
2.3
19.5
13.1
6.6





81
343
IVAVALPALVAP
12
7
50.2
203.3
2.3
19.4
12.9
6.5





76
323
IVAVALPVALAP
12
7
50.2
203.3
2.3
19.1
12.8
6.4





105
461
IAAVIVPAVALP
12
7
50.2
203.3
2.4
19.0
12.7
6.4





9
21
AVALLPALLAVP
12
6
57.3
211.7
2.3
18.9
12.6
6.4





95
404
LAAAVIPAAILP
12
7
54.9
195.8
2.2
18.9
12.6
6.4





60
261
LVLVPLLAAAAP
12
5
41.3
211.6
2.3
18.5
12.3
6.2





122
524
AVALIVVPALAP
12
8
50.2
203.3
2.4
18.3
12.2
6.2





55
225
VAALLPAAAVLP
12
6
57.3
187.5
2.1
18.3
12.2
6.2





63
264
LAAAPVVIVIAP
12
5
50.2
203.3
2.4
18.2
12.1
6.1





1
1
AAALAPVVLALP
12
6
57.3
187.5
2.1
17.7
11.8
6.0





88
382
AAALVIPAILAP
12
7
54.9
195.8
2.2
17.7
11.8
6.0





107
463
AVAILVPLLAAP
12
7
57.3
211.7
2.4
17.6
11.7
5.9





75
322
VVAIVLPALAAP
12
7
50.2
203.3
2.3
17.6
11.7
5.9





117
503
AAIIIVLPAALP
12
8
50.2
220.0
2.4
17.6
11.8
5.9





211
870
VLVAAVLPIAAP
12
8
41.3
203.3
2.4
16.6
11.1
5.6





56
241
AAAVVPVLLVAP
12
6
57.3
195.0
2.4
16.6
11.0
5.6





163
726
LAVAIIAPAVAP
12
8
57.3
187.5
2.2
16.5
11.0
5.6





79
341
IVAVALPAVLAP
12
7
50.2
203.3
2.3
16.4
10.9
5.5





125
542
ALALIIVPAVAP
12
8
50.2
211.6
2.4
16.2
10.8
5.5





83
361
AVVIVAPAVIAP
12
7
50.2
195.0
2.4
16.0
10.7
5.4





54
224
ILAAVPIALAAP
12
6
57.3
195.8
2.2
15.8
10.6
5.3





20
64
AIVALPVAVLAP
12
6
50.2
203.3
2.4
15.8
10.6
5.3





111
482
ILAVAAIPVAVP
12
8
54.9
203.3
2.4
15.8
10.6
5.3





113
484
LAVVLAAPAIVP
12
8
50.2
203.3
2.4
15.6
10.4
5.3





210
868
VLVAAILPAAIP
12
8
54.9
211.7
2.4
14.9
10.0
5.0





124
541
LLALIIAPAAAP
12
8
57.3
204.1
2.1
14.8
9.9
5.0





150
666
AAIAIIAPAIVP
12
8
50.2
195.8
2.3
14.7
9.9
5.0





149
665
LAIVLAAPVAVP
12
8
50.2
203.3
2.3
14.7
9.9
5.0





84
363
AVLAVAPALIVP
12
7
50.2
203.3
2.3
14.7
9.8
4.9





57
242
AALLVPALVAAP
12
6
57.3
187.5
2.1
14.6
9.7
4.9





90
384
VIVAIAPALLAP
12
7
50.2
211.6
2.4
14.0
9.4
4.7





214
877
VAIIAVPAVVAP
12
7
57.3
195.0
2.4
14.0
9.4
4.7





206
863
AAVVLLPIIAAP
12
7
41.3
211.7
2.4
13.8
9.3
4.7





123
525
ALAIVVAPVAVP
12
8
50.2
195.0
2.4
13.8
9.2
4.7





213
875
AIAIVVPAVAVP
12
7
50.2
195.0
2.4
13.8
9.2
4.7





69
285
AIVLLPAAVVAP
12
6
50.2
203.3
2.4
13.3
8.9
4.5





65
281
ALIVLPAAVAVP
12
6
50.2
203.3
2.4
13.3
8.9
4.5





209
867
ALLVVIAPLAAP
12
8
41.3
211.7
2.4
13.2
8.8
4.4





172
766
IVVIAVAPAVAP
12
8
50.2
195.0
2.4
12.9
8.6
4.4





80
342
VIVALAPAVLAP
12
7
50.2
203.3
2.3
12.7
8.5
4.3





217
881
AALIVVPAVAVP
12
7
50.2
195.0
2.4
12.7
8.5
4.3





119
505
AIIIVIAPAAAP
12
8
50.2
195.8
2.3
12.4
8.3
4.2






















TABLE 31













Relative


SEQ

Proline
Rigidity/
Sturctural

Ratio


ID

Position
Flexibility
Feature
Hydropathy
(Fold)

















NO
aMTD
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)
A
B
C




















169
763
VAVLIAVPALAP
12
8
57.3
203.3
2.3
12.3
7.2
4.2





156
687
AILAVALPLLAP
12
8
57.3
220.0
2.3
12.0
7.0
4.1





159
706
IVAVALLPAVAP
12
8
50.2
203.3
2.4
12.0
7.0
4.1





145
643
LALVLAAPAIVP
12
8
50.2
211.6
2.4
11.8
7.9
4.0





66
282
VLAVAPALIVAP
12
6
50.2
203.3
2.4
11.8
7.9
4.0





126
543
LLAALIAPAALP
12
8
57.3
204.1
2.1
11.7
7.8
4.0





78
325
IVAVALPAVALP
12
7
50.2
203.3
2.3
11.7
7.8
4.0





199
846
IAVAVAAPLLVP
12
8
41.3
203.3
2.4
11.7
6.8
4.0





89
383
VIVALAPALLAP
12
7
50.2
211.6
2.3
11.6
7.7
3.9





87
381
VVAIVLPAVAAP
12
7
50.2
195.0
2.4
11.5
7.7
3.9





183
808
LVVLAAAPLAVP
12
8
41.3
203.3
2.3
11.5
7.6
3.9





208
865
AVLVIAVPAIAP
12
8
57.3
203.3
2.5
11.3
7.5
3.8





162
725
IAVLAVAPAVLP
12
8
57.3
203.3
2.3
11.2
7.5
3.8





197
844
VVALLAPLIAAP
12
7
41.3
211.8
2.4
11.2
7.5
3.8





228
897
AVIVPVAIIAAP
12
5
50.2
203.3
2.5
11.2
7.5
3.8





141
605
VIAAVLAPVAVP
12
8
57.3
195.0
2.4
11.0
7.4
3.7





166
744
AAVVIVAPVALP
12
8
50.2
195.0
2.4
11.0
7.3
3.7





51
221
AAILAPIVALAP
12
6
50.2
195.8
2.2
10.9
7.3
3.7





142
622
ALIVLAAPVAVP
12
8
50.2
203.3
2.4
10.6
7.1
3.6





92
401
AALAVIPAAILP
12
7
54.9
195.8
2.2
10.6
7.1
3.6





77
324
IVAVALPAALVP
12
7
50.2
203.3
2.3
10.3
6.9
3.5





215
878
IVALVAPAAVVP
12
7
50.2
195.0
2.4
10.3
6.9
3.5





71
302
LALAPALALLAP
12
5
57.3
204.2
2.1
10.2
6.8
3.4





154
685
ALLVAVLPAALP
12
8
57.3
211.7
2.3
10.2
5.9
3.4





201
848
AVAIVVLPAVAP
12
8
50.2
195.0
2.4
10.0
6.7
3.4





138
602
VIVALAAPVLAP
12
8
50.2
203.3
2.4
9.9
5.8
3.4





178
788
AIAVAIAPVALP
12
8
57.3
187.5
2.3
9.8
6.6
3.3





38
145
LLAVVPAVALAP
12
6
57.3
203.3
2.3
9.5
6.3
3.2





6
11
VVALAPALAALP
12
6
57.3
187.5
2.1
9.5
6.3
3.2





35
141
AVIVLPALAVAP
12
6
50.2
203.3
2.4
9.4
6.3
3.2





120
521
LAALIVVPAVAP
12
8
50.2
203.3
2.4
9.4
6.3
3.2





100
425
AVVAIAPVLALP
12
7
57.3
203.3
2.4
9.4
6.3
3.2





86
365
AVIVVAPALLAP
12
7
50.2
203.3
2.3
9.3
6.2
3.1





62
263
ALAVIPAAAILP
12
6
54.9
195.8
2.2
9.0
6.0
3.0





82
345
ALLIVAPVAVAP
12
7
50.2
203.3
2.3
8.9
5.9
3.0





203
850
LVIALAAPVALP
12
8
57.3
211.7
2.4
8.8
5.9
3.0





37
144
VLAIVPAVALAP
12
6
50.2
203.3
2.4
8.8
5.9
3.0





173
767
IVVAAVVPALAP
12
8
50.2
195.0
2.4
8.5
5.0
2.9





47
185
AALVLPLIIAAP
12
6
41.3
220.0
2.4
8.5
5.7
2.9





202
849
AVILLAPLIAAP
12
7
57.3
220.0
2.4
8.3
4.8
2.8





40
162
AVVALPAALIVP
12
6
50.2
203.3
2.4
8.2
5.5
2.8





207
864
ALLVIAPAIAVP
12
7
57.3
211.7
2.4
8.2
4.8
2.8





42
164
LAAVLPALLAAP
12
6
57.3
195.8
2.1
8.2
5.5
2.8





236
907
VAIALAPVVVAP
12
7
57.3
195.0
2.4
8.1
5.4
2.8





103
444
LAAALVPVALVP
12
7
57.3
203.3
2.3
8.1
5.4
2.7





102
443
ALAALVPVALVP
12
7
57.3
203.3
2.3
8.0
5.3
2.7





221
887
VLAVAPAVAVLP
12
6
57.3
195.0
2.4
7.7
5.1
2.6





231
901
ALVAVLPAVAVP
12
7
57.3
195.0
2.4
7.7
5.1
2.6





167
746
VAIIVVAPALAP
12
8
50.2
203.3
2.4
7.6
4.4
2.6





232
902
ALVAPLLAVAVP
12
5
41.3
203.3
2.3
7.6
5.1
2.6





133
565
VAIVLVAPAVAP
12
8
50.2
195.0
2.4
7.5
5.0
2.5





59
245
AAALAPVLALVP
12
6
57.3
187.5
2.1
7.5
5.0
2.5





165
743
AIAIALVPVALP
12
8
57.3
211.6
2.4
7.4
4.9
2.5





109
465
AVVILVPLAAAP
12
7
57.3
203.3
2.4
7.4
4.9
2.5





30
104
AVVAAPLVLALP
12
6
41.3
203.3
2.3
7.3
4.9
2.5






















TABLE 32













Relative


SEQ

Proline
Rigidity/
Sturctural

Ratio


ID

Position
Flexibility
Feature
Hydropathy
(Fold)

















NO
aMTD
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)
A
B
C




















160
707
IVALAVLPAVAP
12
8
50.2
203.3
2.4
7.3
4.9
2.5





212
872
VLAAAVLPLVVP
12
8
41.3
219.2
2.5
7.3
4.9
2.5





135
583
AVILALAPIVAP
12
8
50.2
211.6
2.4
7.3
4.8
2.4





216
879
AAIVLLPAVVVP
12
7
50.2
219.1
2.5
7.2
4.8
2.4





175
784
VAALPAVALVVP
12
5
57.3
195.0
2.4
7.1
4.7
2.4





225
893
VIAIPAILAAAP
12
5
54.9
195.8
2.3
7.0
4.7
2.4





8
13
AAALVPVVALLP
12
6
57.3
203.3
2.3
7.0
4.7
2.4





184
809
LIVLAAPALAAP
12
7
50.2
195.8
2.2
7.0
4.7
2.4





104
445
ALAALVPALVVP
12
7
57.3
203.3
2.3
6.9
4.6
2.3





22
81
AALLPALAALLP
12
5
57.3
204.2
2.1
6.9
4.6
2.3





151
667
LAVAIVAPALVP
12
8
50.2
203.3
2.3
6.9
4.6
2.3





235
906
AVIALAPVVVAP
12
7
57.3
195.0
2.4
6.8
4.6
2.3





112
483
ILAAAIIPAALP
12
8
54.9
204.1
2.2
6.8
4.5
2.3





114
485
AILAAIVPLAVP
12
8
50.2
211.6
2.4
6.8
4.5
2.3





97
421
AAILAAPLIAVP
12
7
57.3
195.8
2.2
6.7
4.5
2.3





136
585
ALIVAIAPALVP
12
8
50.2
211.6
2.4
6.6
4.4
2.2





99
424
AVVVAAPVLALP
12
7
57.3
195.0
2.4
6.6
4.4
2.2





85
364
LVAAVAPALIVP
12
7
50.2
203.3
2.3
6.5
4.3
2.2





93
402
ALAAVIPAAILP
12
7
54.9
195.8
2.2
6.4
4.3
2.2





106
462
IAAVLVPAVALP
12
7
57.3
203.3
2.4
6.3
4.2
2.1





64
265
VLAIAPLLAAVP
12
6
41.3
211.6
2.3
6.0
4.0
2.0





70
301
VIAAPVLAVLAP
12
6
57.3
203.3
2.4
6.0
4.0
2.0





45
183
LLAAPVVIALAP
12
6
57.3
211.6
2.4
6.0
4.0
2.0





58
243
AAVLLPVALAAP
12
6
57.3
187.5
2.1
5.9
3.9
2.0





148
664
ILIAIAIPAAAP
12
8
54.9
204.1
2.3
5.7
3.8
1.9





174
783
IVALVPAVAIAP
12
6
50.2
203.3
2.5
5.7
3.8
1.9





116
502
AIVALAVPVLAP
12
8
50.2
203.3
2.4
5.6
3.7
1.9





61
262
ALIAVPAIIVAP
12
6
50.2
211.6
2.4
5.5
3.7
1.9





152
683
LAIVLAAPAVLP
12
8
50.2
211.7
2.4
5.5
3.2
1.9





193
830
IALVAAPVALVP
12
7
57.3
203.3
2.4
5.3
3.5
1.8





170
764
AVALAVLPAVVP
12
8
57.3
195.0
2.3
5.0
3.4
1.7





182
807
AVALAVPALVLP
12
7
57.3
203.3
2.3
5.0
3.3
1.7





46
184
LAAIVPAIIAVP
12
6
50.2
211.6
2.4
4.8
3.2
1.6





73
305
IALAAPILLAAP
12
6
57.3
204.2
2.2
4.8
3.2
1.6





27
101
LVALAPVAAVLP
12
6
57.3
203.3
2.3
4.5
3.0
1.5





72
304
AIILAPIAAIAP
12
6
57.3
204.2
2.3
4.4
3.0
1.5





140
604
VALIAVAPAVVP
12
8
57.3
195.0
2.4
4.3
2.5
1.5





146
645
ALAVVALPAIVP
12
8
50.2
203.3
2.4
4.3
2.9
1.5





48
201
LALAVPALAALP
12
6
57.3
195.8
2.1
4.2
2.8
1.4





41
163
LALVLPAALAAP
12
6
57.3
195.8
2.1
4.1
2.4
1.4





195
832
AVAAIVPVIVAP
12
7
43.2
195.0
2.5
4.1
2.7
1.4





44
182
ALIAPVVALVAP
12
6
57.3
203.3
2.4
4.0
2.7
1.4





11
23
VVLVLPAAAAVP
12
6
57.3
195.0
2.4
4.0
2.6
1.3





31
105
LLALAPAALLAP
12
6
57.3
204.1
2.1
4.0
2.6
1.3





129
561
AAVAIVLPAVVP
12
8
50.2
195.0
2.4
3.9
2.6
1.3





171
765
AVALAVVPAVLP
12
8
57.3
195.0
2.3
3.8
2.2
1.3





153
684
AAIVLALPAVLP
12
8
50.2
211.7
2.4
3.5
2.1
1.2





36
143
AVLAVPAVLVAP
12
6
57.3
195.0
2.4
3.3
2.2
1.1





118
504
LIVALAVPALAP
12
8
50.2
211.7
2.4
3.3
2.2
1.1





10
22
AVVLVPVLAAAP
12
6
57.3
195.0
2.4
3.1
2.1
1.1





5
5
AAALLPVALVAP
12
6
57.3
187.5
2.1
3.1
2.1
1.0





67
283
AALLAPALIVAP
12
6
50.2
195.8
2.2
3.1
2.0
1.0





21
65
IAIVAPVVALAP
12
6
50.2
203.3
2.4
3.0
2.0
1.0





219
883
LAIVPAAIAALP
12
6
50.2
195.8
2.2
3.0
2.0
1.0





33
123
AAIIVPAALLAP
12
6
50.2
195.8
2.2
2.9
2.0
1.0






















TABLE 33













Relative


SEQ

Proline
Rigidity/
Sturctural

Ratio


ID

Position
Flexibility
Feature
Hydropathy
(Fold)

















NO
aMTD
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)
A
B
C




















68
284
ALIAPAVALIVP
12
5
50.2
211.7
2.4
2.8
1.8
0.9





50
205
ALALVPAIAALP
12
6
57.3
195.8
2.2
2.6
1.7
0.9





14
42
VAALPVVAVVAP
12
5
57.3
186.7
2.4
2.5
1.7
0.8





32
121
AIVALPALALAP
12
6
50.2
195.8
2.2
2.5
1.7
0.8





13
25
IVAVAPALVALP
12
6
50.2
203.3
2.4
2.4
1.6
0.8





12
24
IALAAPALIVAP
12
6
50.2
195.8
2.2
2.3
1.6
0.8





49
204
LIAALPAVAALP
12
6
57.3
195.8
2.2
2.2
1.5
0.8





7
12
LLAAVPAVLLAP
12
6
57.3
211.7
2.3
2.2
1.5
0.7





15
43
LLAAPLVVAAVP
12
5
41.3
187.5
2.1
2.1
1.4
0.7





29
103
ALIAAPILALAP
12
6
57.3
204.2
2.2
2.1
1.4
0.7





23
82
AVVLAPVAAVLP
12
6
57.3
195.0
2.4
2.1
1.4
0.7





4
4
ALALLPVAALAP
12
6
57.3
195.8
2.1
2.0
1.3
0.7





26
85
LLVLPAAALAAP
12
5
57.3
195.8
2.1
1.9
1.3
0.7





19
63
AALLVPALVAVP
12
6
57.3
203.3
2.3
1.9
1.3
0.7





16
44
ALAVPVALLVAP
12
5
57.3
203.3
2.3
1.6
1.1
0.5





25
84
AAVAAPLLLALP
12
6
41.3
195.8
2.1
1.5
1.0
0.5





18
62
VALLAPVALAVP
12
6
57.3
203.3
2.3
1.4
0.9
0.5





24
83
LAVAAPLALALP
12
6
41.3
195.8
2.1
1.4
0.9
0.5





28
102
LALAPAALALLP
12
5
57.3
204.2
2.1
1.4
0.9
0.5





143
623
VAAAIALPAIVP
12
8
50.2
187.5
2.3
0.8
0.6
0.3













19.6 ± 1.6
13.1 ± 1.1
6.6 ± 0.5









Moreover, compared to reference CPPs (B type: MTM12 and C type: MTD85), novel 240 aMTDs averaged of 13±1.1 (maximum 109.9) and 6.6±0.5 (maximum 55.5) fold higher cell-permeability, respectively (Tables 29 to 33).














TABLE 34








Negative






control






rP38
MTM12
MTD85









aMTD
19.6 ± 1.6*
13.1 ± 1.1*
6.6 ± 0.5*



The Average of
(Best: 164.2)
(Best: 109.9)
(Best: 55.5)



240 aMTDs







*Relative Fold (aMTD in Geo Mean in its comparison to rP38, MTM12, MTD85)






In addition, cell-permeability of 31 rPeptides has been compared with that of 240 aMTDs (0.3±0.04; Tables 35 and 36).

















TABLE 35













Relative






Proline
Rigidity/
Sturctural

Ratio to






Position
Flexibility
Feature
Hydropathy
aMTD


Number
rPeptide
Sequences
Length
(PP)
(II)
(AI)
(GRAVY)
AVE























1
692
PAPLPPVVILAV
12
1, 3, 5, 6
105.5
186.7
1.8
0.74





2
26
AAIALAAPLAIV
12
8
18.1
204.2
2.5
0.65





3
113
PVAVALLIAVPP
12
1, 11, 12
57.3
195
2.1
0.61





4
466
IIAAAAPLAIIP
12
7, 12
22.8
204.2
2.3
0.52





5
167
VAIAIPAALAIP
12
6, 12
20.4
195.8
2.3
0.50





6
97
ALLAAPPALLAL
12
6, 7 
57.3
204.2
2.1
0.41





7
390
VPLLVPVVPVVP
12
2, 6, 9, 12
105.4
210
2.2
0.41





8
426
AAALAIPLAIIP
12
7, 12
4.37
204.2
2.2
0.40





9
214
ALIVAPALMALP
12
6, 12
60.5
187.5
2.2
0.33





10
68
VAPVLPAAPLVP
12
3, 6, 9, 12
105.5
162.5
1.6
0.32





11
39
CYNTSPCTGCCY
12
6
52.5
0.0
0.0
0.29





12
934
LILAPAAVVAAA
12
5
57.3
195.8
2.5
0.28





13
938
VPVLLPVVVPVP
12
2, 6, 10, 12
121.5
210
2.2
0.28





14
329
LPVLVPVVPVVP
12
2, 6, 9, 12
121.5
210
2.2
0.23





15
606
AAAIAAIPIIIP
12
8, 12
4.4
204.2
2.4
0.20





16
49
VVPAAPAVPVVP
12
3, 6, 9, 12
121.5
145.8
1.7
0.18





17
139
TGSTNSPTCTST
12
7
53.4
0.0
−0.7
0.17





18
772
LPVAPVIPIIVP
12
2, 5, 8, 12
79.9
210.8
2.1
0.16





19
921
IWWFVVLPLVVP
12
8, 12
41.3
194.2
2.2
0.14





20
66
AGVLGGPIMGVP
12
7, 12
35.5
121.7
1.3
0.13





21
693
AAPVLPVAVPIV
12
3, 6, 10
82.3
186.7
2.1
0.13





22
18
NYCCTPTTNGQS
12
6
47.9
0.0
−0.9
0.10





23
16
NNSCTTYTNGSQ
12
None
47.4
0.0
−1.4
0.08





24
227
LAAIVPIAAAVP
12
6, 12
34.2
187.5
2.2
0.08





25
17
GGCSAPQTTCSN
12
6
51.6
8.3
−0.5
0.08





26
67
LDAEVPLADDVP
12
6, 12
34.2
130
0.3
0.08





27
635
GSTGGSQQNNQY
12
None
31.9
0.0
−1.9
0.07





28
29
VLPPLPVLPVLP
12
3, 4, 6, 9, 12
121.5
202.5
1.7
0.07





29
57
QNNCNTSSQGGG
12
None
52.4
0.0
−1.6
0.06





30
700
GTSNTCQSNQNS
12
None
19.1
0.0
−1.6
0.05





31
38
YYNQSTCGGQCY
12
None
53.8
0.0
−1.0
0.05












AVE
0.3 ± 0.04



















TABLE 36








Relative Ratio




to aMTD AVE*









rPeptide
0.3 ± 0.04



The Average




of 31 aMTDs







*Out of 240 aMTDs, average relative fold of aMTD had been 19.6 fold compared to type A (rP38).






In summary, relative cell-permeability of aMTDs has shown maximum of 164.0, 109.9 and 55.5 fold higher to rP38, MTM12 and MTD85, respectively. In average of total 240 aMTD sequences, 19.6±1.6, 13.1±1.1 and 6.6±0.5 fold higher cell-permeability are shown to the rP38, MTM12 and MTD85, respectively (Tables 29 to 33). Relative cell-permeability of negative control (rP38) to the 240 aMTDs is only 0.3±0.04 fold.


4-5. Intracellular Delivery and Localization of aMTD-Fused Recombinant Proteins


Recombinant proteins fused to the aMTDs were tested to determine their intracellular delivery and localization by laser scanning confocal microscopy with a negative control (rP38) and previous published CPPs (MTM12 and MTD85) as the positive control references. NIH3T3 cells were exposed to 10 μM of FITC-labeled protein for 1 hour at 37, and nuclei were counterstained with DAPI. Then, cells were examined by confocal laser scanning microscopy (FIG. 7). Recombinant proteins fused to aMTDs clearly display intracellular delivery and cytoplasmic localization (FIG. 7) that are typically higher than the reference CPPs (MTM12 and MTD85). The rP38-fused recombinant protein did not show internalized fluorescence signal (FIG. 7a). In addition, as seen in FIG. 8, rPeptides (his-tagged CRA recombinant proteins fused to each rPeptide) display lower- or non-cell-permeability.


4-6. Summary of Quantitative and Visual Cell-Permeability of Newly Developed aMTDs


Histidine-tagged aMTD-fused cargo recombinant proteins have been greatly enhanced in their solubility and yield. Thus, FITC-conjugated recombinant proteins have also been tested to quantitate and visualize intracellular localization of the proteins and demonstrated higher cell-permeability compared to the reference CPPs.


In the previous studies using the hydrophobic signal-sequence-derived CPPs—MTS/MTM or MTDs, 17 published sequences have been identified and analyzed in various characteristics such as length, molecular weight, pI value, bending potential, rigidity, flexibility, structural feature, hydropathy, amino acid residue and composition, and secondary structure of the peptides. Based on these analytical data of the sequences, novel artificial and non-natural peptide sequences designated as advanced MTDs (aMTDs) have been invented and determined their functional activity in intracellular delivery potential with aMTD-fused recombinant proteins.


aMTD-fused recombinant proteins have promoted the ability of protein transduction into the cells compared to the recombinant proteins containing rPeptides and/or reference hydrophobic CPPs (MTM12 and MTD85). According to the results, it has been demonstrated that critical factors of cell-penetrating peptide sequences play a major role to determine peptide-mediated intracellular delivery by penetrating plasma membrane. In addition, cell-permeability can considerably be improved by following the rational that all satisfy the critical factors.


5. Structure/Sequence Activity Relationship (SAR) of aMTDs on Delivery Potential

After determining the cell-permeability of novel aMTDs, structure/sequence activity relationship (SAR) has been analyzed for each critical factor in selected some of and all of novel aMTDs (FIGS. 13 to 16 and Table 37).














TABLE 37







Rank of
Rigidity/
Sturctural

Relative
Amino Acid


Delivery
Flexibility
Feature
Hydropathy
Ratio (Fold)
Composition

















Potential
(II)
(AI)
(GRAVY)
A
B
C
A
V
I
L




















 1~10
55.9
199.2
2.3
112.7
75.5
38.1
4.0
3.5
0.4
2.1


11~20
51.2
205.8
2.4
56.2
37.6
19.0
4.0
2.7
1.7
1.6


21~30
49.1
199.2
2.3
43.6
28.9
14.6
4.3
2.7
1.4
1.6


31~40
52.7
201.0
2.4
34.8
23.3
11.8
4.2
2.7
1.5
1.6


41~50
53.8
201.9
2.3
30.0
20.0
10.1
4.3
2.3
1.1
2.3


51~60
51.5
205.2
2.4
23.5
15.7
7.9
4.4
2.1
1.5
2.0


222~231
52.2
197.2
2.3
2.2
1.5
0.8
4.5
2.1
1.0
2.4


232~241
54.1
199.7
2.2
1.7
1.2
0.6
4.6
1.7
0.2
3.5









5-1. Proline Position

In regards to the bending potential (proline position: PP), aMTDs with its proline at 7′ or 8′ amino acid in their sequences have much higher cell-permeability compared to the sequences in which their proline position is at 5′ or 6′ (FIGS. 14a and 15a).


5-2. Hydropathy

In addition, when the aMTDs have GRAVY (Grand Average of Hydropathy) ranging in 2.1-2.2, these sequences display relatively lower cell-permeability, while the aMTDs with 2.3-2.6 GRAVY are shown significantly higher one (FIGS. 14b and 15b).


5-3. rPeptide SAR


To the SAR of aMTDs, rPeptides have shown similar SAR correlations in the cell-permeability, pertaining to their proline position (PP) and hydropathy (GRAVY). These results confirms that rPeptides with high GRAVY (2.4 to 2.6) have better cell-permeability (FIG. 16).


5-4. Analysis of Amino Acid Composition

In addition to proline position and hydropathy, the difference of amino acid composition is also analyzed. Since aMTDs are designed based on critical factors, each aMTD-fused recombinant protein has equally two proline sequences in the composition. Other hydrophobic and aliphatic amino acids—alanine, isoleucine, leucine and valine—are combined to form the rest of aMTD peptide sequences.


Alanine: In the composition of amino acids, the result does not show a significant difference by the number of alanine in terms of the aMTD's delivery potential because all of the aMTDs have three to five alanines. In the sequences, however, four alanine compositions show the most effective delivery potential (geometric mean) (FIG. 13a).


Leucine and Isoleucine: Also, the compositions of isoleucine and leucine in the aMTD sequences show inverse relationship between the number of amino acid (I and L) and delivery potential of aMTDs. Lower number of isoleucine and leucine in the sequences tends to have higher delivery potential (geometric mean) (FIGS. 13b and 13c).


Valine: Conversely, the composition of valine of aMTD sequences shows positive correlation with their cell-permeability. When the number of valine in the sequence is low, the delivery potential of aMTD is also relatively low (FIG. 13d).


Ten aMTDs having the highest cell-permeability are selected (average geometric mean: 2584±126). Their average number of valine in the sequences is 3.5; 10 aMTDs having relatively low cell-permeability (average geometric mean: 80±4) had average of 1.9 valine amino acids. The average number of valine in the sequences is lowered as their cell-permeability is also lowered as shown in FIG. 13d. Compared to higher cell-permeable aMTDs group, lower sequences had average of 1.9 in their valine composition. Therefore, to obtain high cell-permeable sequence, an average of 2-4 valines should be composed in the sequence.


5-5. Conclusion of SAR Analysis:

As seen in FIG. 15, all 240 aMTDs have been examined for these association of the cell-permeability and the critical factors: bending potential (PP), rigidity/flexibility (II), structure feature (AI), and hydropathy (GRAVY), amino acid length and composition. Through this analysis, cell-permeability of aMTDs tends to be lower when their central proline position is at 5′ or 6′ and GRAVY is 2.1 or lower (FIG. 15). Moreover, after investigating 10 higher and 10 lower cell-permeable aMTDs, these trends are clearly shown to confirm the association of cell-permeability with the central proline position and hydropathy.


6. Experimental Confirmation of Index Range/Feature of Critical Factors

The range and feature of five out of six critical factors have been empirically and experimentally determined that are also included in the index range and feature of the critical factors initially proposed before conducting the experiments and SAR analysis. In terms of index range and feature of critical factors of newly developed 240 aMTDs, the bending potential (proline position: PP), rigidity/flexibility (Instability Index: II), structural feature (Aliphatic Index: AI), hydropathy (GRAVY), amino acid length and composition are all within the characteristics of the critical factors derived from analysis of reference hydrophobic CPPs.


Therefore, our hypothesis to design and develop new hydrophobic CPP sequences as advanced MTDs is empirically and experimentally proved and demonstrated that critical factor-based new aMTD rational design is correct.









TABLE 38







Summarized Critical Factors of aMTD










Newly
Analysis of



Designed CPPs
Experimental Results


Critical Factor
Range
Range





Bending Potential
Proline presences
Proline presences


(Proline Position: PP)
in the middle
in the middle



(5’, 6’, 7’ or 8’)
(5’, 6’, 7’ or 8’)



and at the
and at the



end of peptides
end of peptides


Rigidity/Flexibility
40-60
41.3-57.3


(Instability Index: II)




Structural Feature
180-220
187.5-220.0


(Aliphatic Index: AI)




Hydropathy
2.1-2.6
2.2-2.6


(Grand Average of




Hydropathy GRAVY)




Length
 9-13
12


(Number of Amino Acid)




Amino acid Composition
A, V, I, L, P
A, V, I, L, P









7. Discovery and Development of Protein-Based New Biotherapeutics with MITT Enabled by aMTDs for Protein Therapy

Total of 240 aMTD sequences have been designed and developed based on the critical factors. Quantitative and visual cell-permeability of 240 aMTDs (hydrophobic, flexible, bending, aliphatic and 12 a/a-length peptides) are all practically determined.


To measure the cell-permeability of aMTDs, rPeptides have also been designed and tested. As seen in FIGS. 13 to 15, there are vivid association of cell-permeability and the critical factors of the peptides. Out of these critical factors, we are able to configure that the most effective cell-permeable aMTDs have the amino acid length of 12; composition of A, V, L, I and P; multiple proline located at either 7′ or 8′ and at the end (12′); instability index ranged of 41.3 to 57.3; aliphatic index ranged of 187.5 to 220.0; and hydropathy (GRAVY) ranged of 2.2 to 2.6.


These examined critical factors are within the range that we have set for our critical factors; therefore, we are able to confirm that the aMTDs that satisfy these critical factors have relatively high cell-permeability and much higher intracellular delivery potential compared to reference hydrophobic CPPs reported during the past two decades.


It has been widely evident that many human diseases are caused by proteins with deficiency or over-expression that causes mutations such as gain-of-function or loss-of-function. If biologically active proteins could be delivered for replacing abnormal proteins within a short time frame, possibly within an hour or two, in a quantitative manner, the dosage may be regulated depending on when and how proteins may be needed. By significantly improving the solubility and yield of novel aMTD in this invention (Table 34), one could expect its practical potential as an agent to effectively deliver therapeutic macromolecules such as proteins, peptides, nucleic acids, and other chemical compounds into live cells as well as live mammals including human. Therefore, newly developed MITT utilizing the pool (240) of novel aMTDs can be used as a platform technology for discovery and development of protein-based biotherapeutics to apprehend intracellular protein therapy after determining the optimal cargo-aMTD relationship.


The following examples are presented to aid practitioners of the invention, to provide experimental support for the invention, and to provide model protocols. In no way are these examples to be understood to limit the invention.


EXAMPLE 1
Development of Novel Advanced Macromolecule Transduction Domain (aMTD)

H-regions of signal sequences (HRSP)-derived CPPs (MTS/MTM and MTD) do not have a common sequence, a sequence motif, and/or a common structural homologous feature. In this invention, the aim is to develop improved hydrophobic CPPs formatted in the common sequence and structural motif that satisfy newly determined ‘critical factors’ to have a ‘common function,’ to facilitate protein translocation across the plasma membrane with similar mechanism to the analyzed CPPs.


The structural motif as follows:




embedded image


In Table 12, universal common sequence/structural motif is provided as follows. The amino acid length of the peptides in this invention ranges from 9 to 13 amino acids, mostly 12 amino acids, and their bending potentials are dependent with the presence and location of proline in the middle of sequence (at 5′, 6′, 7′ or 8′ amino acid) and at the end of peptide (at 12′) for recombinant protein bending. Instability index (II) for rigidity/flexibility of aMTDs is II<40, grand average of hydropathy (GRAVY) for hydropathy is around 2.2, and aliphatic index (AI) for structural features is around 200 (Table 12). Based on these standardized critical factors, new hydrophobic peptide sequences, namely advanced macromolecule transduction domain peptides (aMTDs), in this invention have been developed and summarized in Tables 13 to 18.


EXAMPLE 2
Construction of Expression Vectors for Recombinant Proteins Fused to aMTDs

Our newly developed technology has enabled us to expand the method for making cell-permeable recombinant proteins. The expression vectors were designed for histidine-tagged CRA proteins fused with aMTDs or rPeptides. To construct expression vectors for recombinant proteins, polymerase chain reaction (PCR) had been devised to amplify each designed aMTD or rPeptide fused to CRA.


The PCR reactions (100 ng of genomic DNA, 10 pmol of each primer, each 0.2 mM dNTP mixture, 1× reaction buffer and 2.5 U of Pfu(+) DNA polymerase (Doctor protein, Korea) was digested on the restriction enzyme site between Nde I (5′) and Sal I (3′) involving 35 cycles of denaturation (95° C.), annealing (62° C.), and extension (72° C.) for 30 seconds each. For the last extension cycle, the PCR reactions remained for 5 minutes at 72° C. Then, they were cloned into the site of pET-28a(+) vectors (Novagen, Darmstadt, Germany). DNA ligation was performed using T4 DNA ligase at 4° C. overnight. These plasmids were mixed with competent cells of E. coli DH5-alpha strain on the ice for 10 minutes. This mixture was placed on the ice for 2 minutes after it was heat shocked in the water bath at 42° C. for 90 seconds. Then, the mixture added with LB broth media was recovered in 37° C. shaking incubator for 1 hour. Transformant was plated on LB broth agar plate with kanamycin (50 μg/mL) (Biopure, Johnson City, Tenn., USA) before incubating at 37° C. overnight. From a single colony, plasmid DNA was extracted, and after the digestion of Nde I and Sal I restriction enzymes, digested DNA was confirmed at 645 bp by using 1.2% agarose gels electrophoresis (FIG. 2).


EXAMPLE 3
Inducible Expression, Purification and Preparation of Recombinant Proteins Fused to aMTDs and rPeptides

To express recombinant proteins, pET-28a(+) vectors for the expression of CRA proteins fused to a negative control [rPeptide 38 (rP38)], reference hydrophobic CPPs (MTM12 and MTD85) and aMTDs were transformed in E. coli BL21 (DE3) strains. Cells were grown at 37° C. in LB medium containing kanamycin (50 μg/ml) with a vigorous shaking and induced at OD600=0.6 by adding 0.7 mM IPTG (Biopure) for 2 hours at 37° C. . Induced recombinant proteins were loaded on 15% SDS-PAGE gel and stained with Coomassie Brilliant Blue (InstantBlue, Expedeon, Novexin, UK) (FIG. 3).


The E. coli cultures were harvested by centrifugation at 5,000×rpm for 10 minutes, and the supernatant was discarded. The pellet was re-suspended in the lysis buffer (50 mM NaH2PO4, 10 mM Imidazol, 300 mM NaCl, pH 8.0). The cell lysates were sonicated on ice using a sonicator (Sonics and Materials, Inc., Newtown, Conn., USA) equipped with a probe. After centrifuging the cell lysates at 5,000×rpm for 10 minutes to pellet the cellular debris, the supernatant was incubated with lysis buffer-equilibrated Ni-NTA resin (Qiagen, Hilden, Germany) gently by open-column system (Bio-rad, Hercules, Calif., USA). After washing protein-bound resin with 200 ml wash buffer (50 mM NaH2PO4, 20 mM Imidazol, 300 mM NaCl, pH 8.0), the bounded proteins were eluted with elution buffer (50 mM NaH2PO4, 250 mM Imidazol, 300 mM NaCl, pH 8.0).


Recombinant proteins purified under natural condition were analyzed on 15% SDS-PAGE gel and stained with Coomassie Brilliant Blue (FIG. 4). All of the recombinant proteins were dialyzed for 8 hours and overnight against physiological buffer, a 1:1 mixture of cell culture medium (Dulbecco's Modified Eagle's Medium: DMEM, Hyclone, Logan, Utah, USA) and Dulbecco's phosphate buffered saline (DPBS, Gibco, Grand Island, N.Y., USA). From 316 aMTDs and 141 rPeptides cloned, 240 aMTD- and 31 rPeptide-fused recombinant proteins were induced, purified, prepared and analyzed for their cell-permeability.


EXAMPLE 4
Determination of Quantitative Cell-Permeability of Recombinant Proteins

For quantitative cell-permeability, the aMTD- or rPeptide-fused recombinant proteins were conjugated to fluorescein isothiocyanate (FITC) according to the manufacturer's instructions (Sigma-Aldrich, St. Louis, Mo., USA). RAW 264.7 cells were treated with 10 μM FITC-labeled recombinant proteins for 1 hour at 37° C., washed three times with cold PBS, treated with 0.25% tripsin/EDTA (Sigma-Aldrich, St. Louis, Mo.) for 20 minutes at 37° C. to remove cell-surface bound proteins. Cell-permeability of these recombinant proteins were analyzed by flow cytometry (Guava, Millipore, Darmstadt, Germany) using the FlowJo cytometric analysis software (FIGS. 5 to 6). The relative cell-permeability of aMTDs were measured and compared with the negative control (rP38) and reference hydrophobic CPPs (MTM12 and MTD85) (Table 34).


EXAMPLE 5
Determination of Cell-Permeability and Intracellular Localization of Recombinant Proteins

For a visual reference of cell-permeability, NIH3T3 cells were cultured for 24 hours on coverslip in 24-wells chamber slides, treated with 10 μM FITC-conjugated recombinant proteins for 1 hour at 37° C., and washed three times with cold PBS. Treated cells were fixed in 4% paraformaldehyde (PFA, Junsei, Tokyo, Japan) for 10 minutes at room temperature, washed three times with PBS, and mounted with VECTASHIELD Mounting Medium (Vector laboratories, Burlingame, Calif., USA), and counter stained with DAPI (4′,6-diamidino-2-phenylindole). The intracellular localization of the fluorescent signal was determined by confocal laser scanning microscopy (LSM700, Zeiss, Germany; FIGS. 7 and 8).


EXAMPLE 6
Expression of Cas9 Recombinant Proteins
6-1. Construction of Expression Vectors for Recombinant Proteins

Our newly developed technology, aMTD-based MITT, has enabled us to improve the method for developing cell-permeable recombinant proteins. The expression vectors were designed for Cas9 recombinant proteins fused with aMTD/SDs (HNM563C9, HNM563C9SB, HNM563C9SBSB, HNSBC9M563, HNSBSBC9M563) and control proteins without aMTD (HC9). To acquire expression vectors for Cas9 recombinant proteins, polymerase chain reaction (PCR) had been devised to amplify these recombinant proteins.


The PCR reactions (100 ng of genomic DNA, 10 pmol of each primer, each 0.2 mM dNTP mixture, 1× reaction buffer and 2.5 U of Pfu(+) DNA polymerase (Doctor Protein, Korea)) was digested on the different restriction enzyme site involving 40 cycles of denaturation (95 V) for 45 seconds, annealing (58 V) for 45 seconds, and extension (72° C.) for 120 seconds. For the last extension cycle, the PCR reactions remained for 10 minutes at 72° C.


Histidine-tagged Cas9 recombinant proteins are constructed by amplifying the Cas9 cDNA (1368 amino acids) from nt (nucleotide) 1 to 4104, using the primers (Table 39), for aMTD/SD-fused to Cas9 cargo. NLS/aMTD-Cas9 and SDB are prepared by amplifying its templates using the primers. The PCR products of NLS/aMTD-Cas9 and SDB are cleaved with NdeI/BamHI and BamHI/SalI, respectively. The amplified and cohesive-ended SDB are ligated to the BamHI site of the C-terminus of Cas9, then finally ligated into 6xHis expression vector, pET-28a(+) (Novagen, Mdison, Wisc., USA). In addition, Cas9 and NLS/aMTD-Cas9 are amplified its template using the primers (Tables 38 and 39). The PCR products of Cas9 and NLS/aMTD-Cas9 are cleaved with NdeI/BamHI. The amplified and cohesive-ended Cas9 and NLS/aMTD-Cas9 were ligated to the NdeI/BamHI site of the pET-28a(+) vector. DNA ligation was performed using T4 DNA ligase (NEB, USA) at 4° C. overnight. These plasmids were mixed with competent cells of E.coli BL21(DE3) CodonPlus-RIL strain (ATCC, USA) on the ice for 10 minutes. This mixture was placed on the ice for 2 minutes after it was heat-shocked in the water bath at 42° C. for 90 seconds. Then, the mixture added with LB broth media (ELPIS, Korea) was recovered in 37° C. shaking incubator for 1 hour. Then, transformant was plated on LB broth agar plate with kanamycin (25 ug/mL) (Biopure, Johnson City, Tenn.) before incubating overnight at 37° C. From a single colony, plasmid DNA was extracted; and after the double digestion of Ndel and Sall restriction enzymes, digested DNA was confirmed by using 1.2% agarose gels electrophoresis (FIG. 18).


The amino acid and polynucleotide sequences of histidine tag are indicated in SEQ ID NOs: 1217 and 1218; and amino acid and polynucleotide sequences of aMTDs are indicated in SEQ ID NOs: 131 and 371, respectively. The amino acid and polynucleotide sequences of NLS are indicated in SEQ ID NOs: 1219 and 1220, The amino acid and polynucleotide sequences of Cas9 are indicated in SEQ ID NOs: 1221 and 1222 respectively.


As shown in FIG. 18, it was confirmed that the Cas9 recombinant proteins (HNM563C9SB) were expressed from the respective recombinant expression vectors.


PCR primers for the His-tagged Cas9 recombinant proteins fused to aMTD and SD were summarized in Tables 39 and 40 (SEQ ID NOs: 1223 to 1229).













TABLE 39









SEQ


Cargo
SD
Recombinant
5′ Primer (5′ > 3′)
ID NO







Cas9

HC9
CCCAAGCTTATCAACGGTATTCGTGACAAACAAAGC
1223




HNM563C9
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGGCGC
1224




HNM563C9SB
TGGCGGTGATTGTGGTGCCGGCGCTGGCGCCGATGGAC





AAAAAGTATAGCATTGGCCTG



SDB
HNM563C9SB
CCCGGATCCATGGCAGAACAAAGCGACAAGGATGTGAAG
1225




















TABLE 40









SEQ


Cargo
SD
Recombinant
3′ primer (5′ >3′)
ID NO







Cas9

HC9
CCCAAGCTTACGGCTCAGACGGCCCCAACCGGTGTA
1226




HNM563C9
CGCGGATTCATCGCCGCCCAGTTGGCTCAGGTCAAT
1227




HNM563C9SB
CGCGGATTCTTAATCGCCGCCCAGTTGGCTCAGGTCAAT
1228



SDB
HNM563C9SB
CGCGTCGACTTAAAGGGTTTCCGAAGGCTTGGCTATCTT
1229









6-2. Expression and Purification of Histidine-Tagged Cas9 Recombinant Proteins

The transformant was cultured in TB medium containing 25 ug/ml of kanamycin, and the transformant was inoculated in 7 ml of TB medium at 37° C. overnight. The incubated transformant was inoculated in 700 ml of TB medium at 37° C. until OD600 reached 0.4. The medium was added with 0.3 mM isopropyl-β-D-thiogalactoside (IPTG) as a protein expression inducer, and further incubated at 18° C. for 16 hours. The medium was centrifuged at 4° C. and 8,000×g for 5 minutes, and a supernatant was discarded to recover a cell pellet. The pellet was loaded on SDS-PAGE to analyze expression levels. The pellet was suspended in a lysis buffer (20 mM Tris-HCl, pH 7.4, 300 mM KCl, 10% glycerol, 1% sucrose), and then this suspension was disrupted with sonication to the cells. The disrupted cells were centrifuged at 4° C. and 15,000×g for 30 minutes to obtain a soluble fraction and an insoluble fraction. After, the soluble fraction was used for protein purification. Recombinant proteins are supposed to be purified by Ni2+ affinity chromatography as directed by the supplier (Qiagen, Hilden, Germany) in the natural condition. After purification, they will be changed to a physiological buffer, 20 mM Tris, pH 7.4, 300 mM KCl, 20% glycerol, 1% sucrose.


6-3. Determination of Solubility/Yield of Cas9 Recombinant Proteins

The aMTD-fused Cas9 recombinant proteins containing SDB are cloned, expressed, purified, and prepared in a soluble form under the native condition. Each recombinant protein; HNM563C9SB and HNM563C9SBSB were determined for their size (number of amino acids), yield (mg/L) and solubility on 8% SDS-PAGE gel and stained with Coomassie Brilliant Blue.


Solubility will be scored on a 5-point scale ranging from highly soluble proteins with little tendency to precipitate (+++++) to largely insoluble proteins (+) by measuring their turbidity (A450). Yield (mg/L) in physiological buffer condition of each recombinant protein will also be determined.


As shown in FIG. 19, the purified Cas9 recombinant proteins were observed as a single band, where the amount of the final purified protein was up to 38.5 mg/L. It was also confirmed that HNM563C9SB showed excellent yield and solubility.


Then, HNM563C9SB was used as a structure of CP-Cas9 recombinant protein in the next examples.


6-4. Determinaclation of Optimal aMTD for CP-Cas9 Recombinant Proteins


To improve solubility/yield, cell/tissue-permeability and biological activity of the CP-Cas 9 recombinant protein, CP-Cas9 recombinant proteins fused with different aMTDs were prepared (FIG. 20). From 240 aMTDs, 9 aMTDs were selected and used for th e construction of CP-Cas9 recombinant proteins. 9 aMTDs used are shown in the following Table 41 to 43.












TABLE 41









Bending Potential














aMTD
Amino Acid
Proline Position

Felxibility
Feature
Hydropathy


















ID
Sequence
Length
5′
6′
7′
8′
12′
Number
(II)
(AI)
(GRAVITY)





161
ALAVIVVPALAP
12
0
0
0
1
1
12
57.3
195.8
2.2





165
ALAVPVALAIVP
12
1
0
0
0
1
12
50.2
203.3
2.4





264
LAAAPVVIVIAP
12
1
0
0
0
1
12
50.2
203.3
2.4





442
ALAALVPAVLVP
12
0
0
1
0
1
12
57.3
203.3
2.3





522
ALLVIAVPAVAP
12
0
0
0
1
1
12
57.3
203.3
2.4





563
ALAVIVVPALAP
12
0
0
0
1
1
12
50.2
203.3
2.4





661
AAILAPIVAALP
12
0
1
0
0
1
12
50.2
195.8
2.2





889
ILVAAAPIAALP
12
0
0
1
0
1
12
57.3
195.8
2.2





899
AVVIALPAVVAP
12
0
0
1
0
1
12
57.3
195.0
2.4


















TABLE 42





aMTD ID
Amino Acid Sequence
cDNA Sequence







161
ALAVIVVPALAP
GCGCTGGCGGTGATTGTGGTGCCGGCGCTGGCGCCG





165
ALAVPVALAIVP
GCGCTGGCGGTGCCGGTGGCGCTGGCGATTGTGCCG





264
LAAAPVVIVIAP
CTGGCGGCGGCGCCGGTGGTGATTGTGATTGCGCCG





442
ALAALVPAVLVP
GCGCTGGCGGCGCTGGTTCCGGCGGTTCTGGTTCCG





522
ALLVIAVPAVAP
GCGCTGCTGGTGATTGCGGTTCCGGCGGTGGCGCCG





563
ALAVIVVPALAP
CGGCGCCAGCGCCGGCACCACAATCACCGCCAGCGC





661
AAILAPIVAALP
GCGGCGATTCTGGCGCCGATTGTGGCGGCGCTGCCG





889
ILVAAAPIAALP
ATTCTGGTGGCGGCGGCGCCGATTGCGGCGCTGCCG





899
AVVIALPAVVAP
GCGGTGGTGATTGCGCTGCCGGCGGTGGTGGCGCCG



















TABLE 43








SEQ



Recombinant

ID


Cargo
Protein
5′ Primer (5′->3′)
NO







Cas9
HNM161C9SB
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGGCGCTGGCGGTGATT
1234




GTGGTGCCGGCGCTGGCGCCGATGGACAAAAAGTATAGCATTGGCCTG



HNM165C9SB
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGGCGCTGGCGGTGCCG
1235




GTGGCGCTGGCGATTGTGCCGATGGACAAAAAGTATAGCATTGGCCTG



HNM264C9SB
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGCTGGCGGCGGCGCCG
1236




GTGGTGATTGTGATTGCGCCGATGGACAAAAAGTATAGCATTGGCCTG



HNM442C9SB
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGGCGCTGGCGGCGCTG
1237




GTTCCGGCGGTTCTGGTTCCGATGGACAAAAAGTATAGCATTGGCCTG



HNM552C9SB
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGGCGCTGCTGGTGATTG
1238




CGGTTCCGGCGGTGGCGCCGATGGACAAAAAGTATAGCATTGGCCTG



HNM563C9SB
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGGCGCTGGCGGTGATT
1239




GTGGTGCCGGCGCTGGCGCCGATGGACAAAAAGTATAGCATTGGCCTG



HNM661C9SB
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGGCGGCGATTCTGGCG
1240




CCGATTGTGGCGGCGCTGCCGATGGACAAAAAGTATAGCATTGGCCTG



HNM889C9SB
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGATTCTGGTGGCGGCG
1241




GCGCCGATTGCGGCGCTGCCGATGGACAAAAAGTATAGCATTGGCCTG



HNM899C9SB
GGAATTCCATATGCCCAAGAAGAAGAGGAAGGTGGCGGTGGTGATTGCG
1242




CTGCCGGCGGTGGTGGCGCCGATGGACAAAAAGTATAGCATTGGCCTG


















TABLE 44







SEQ




ID



3′ Primer (5′->3′
NO







Cas9
CGCGGATTCTTAATCGCCGCCCAGTTGGCTCAG
1243


Recombinant
GTCAAT


Protein









All of the CP-Cas9 recombinant proteins fused with various aMTDs have excellent yield and solubility.


EXAMPLE 7
Determination of Cell Permeability of CP-Cas9 Recombinant Proteins

To examine cell permeability of CP-Cas9 recombinant protein, CP-Cas9 recombinant proteins were conjugated to 5/6-fluorescein isothiocyanate (FITC). RAW 264.7 (KCLB, Seoul, South Korea) or HeLa cells (KCLB, Seoul, South Korea) were treated with 5 μM FITC-labeled CP-Cas9 recombinant proteins and cultivated at 37° C.


First, RAW 264.7 cells were cultured in a DMEM medium containing 10% fetal bovine serum (FBS, Hyclone, USA) and 1% penicillin/streptomycin (Hyclone, USA). After cultivation, the cells were washed three times with ice-cold PBS (Phosphate-buffered saline, Hyclone, USA) and treated with proteinase K (10 μg/mL, SIGMA, USA) to remove surface-bound proteins, and internalized proteins were measured by flow cytometry (FlowJo cytometric analysis software, Guava, Millipore, Darmstadt, Germany). As a control, untreated cells (vehicle) and equimolar concentration of unconjugated FITC-treated cells (FITC) were used.


Next, HeLa cells was incubated for 3 hour at 37° C. with 5 μM FITC-labeled CP-Cas9 recombinant protein. For nuclear staining, a mixture of VECTASHIELD Mounting Medium (Vector laboratories, Burlingame, Calif.) and DAPI (4′,6-diamidino-2-phenylindole) was added to HeLa cells, and visualized using a confocal laser microscope (LSM700, Zeiss, Germany).


As shown in FIGS. 21 and 22, aMTD563-fused Cas9 (CP-Cas9) recombinant proteins were observed in nucleus of the cells. As a result, CP-Cas9 recombinant protein has excellent cell permeability.


EXAMPLE 8
Determination of Biological Activity of CP-Cas9 Recombinant Proteins

To investigate the biological activity of CP-Cas9 recombinant protein, in vitro plasmid cleavage assay was performed and nuclease activity was measured.


8-1. Nuclease Activity

The mixture was mixed with 250 ng of CP-Cas9 recombinant protein and 50 ng of gRNA (5′-AGGCAAAATGCCGCAAAAAA-3′), and incubated with 300 ng of pUC19 plasmid for 1 hour at 37° C. in 10 μl of reaction buffer (100 mM NaCl, 50 mM Tris-HCl, 10 mM MgCl2 and 100 m/ml of BSA pH 7.9).


gRNA was synthesized using a GeneArt™ Precision gRNA Synthesis kit (Thermo Fisher Scientific, Calif., USA) and recognized part or all of the ampicillin sequence of the plasmid as a target sequence. The target sequence has a nucleotide sequence selected from the group consisting of 5′-TGCCGCAAAAAAGGGAATAA-3′, 5′ -ATGCCGCAAAAAAGGGAATA-3′, 5′-AGGCAAAATGCCGCAAAAAA-3′. Preferably, the target sequence has 5′-AGGCAAAATGCCGCAAAAAA-3′.


When the CP-Cas9 recombinant protein and gRNA are treated in pUC19 plasmid, the plasmid is cleaved by the CP-Cas9 recombinant protein and linearized plasmid is detected. The pUC19 plasmid (Circular) and its linear cleavage product (linear) were separated by 1% agarose gel electrophoresis.


As shown in FIG. 23, the CP-Cas9 recombinant protein has an excellent biological activity as nuclease.


8-2. Nuclease Activity in a Dose Dependent Manner of gRNA


To investigate the gRNA dose dependent nuclease activity of CP-Cas9 recombinant protein, the nuclease activity were measured in the same manner as in Example 8-1.


The mixture was mixed with 200 ng of CP-Cas9 recombinant proteins and 6, 12.5, 25, 50 or 100 ng of gRNA, and incubated with 300 ng of pUC19 plasmid for 1 hour at 37° C. in 10 μl of reaction buffer.


As a positive control, the pUC19 plasmid was treated with 3.1 buffer.


As shown in FIG. 24, the CP-Cas9 recombinant protein has an excellent nuclease activity at all gRNA dose.


8-3. Nuclease Activity in a Dose Dependent Manner of CP-Cas9 Recombinant Protein

To investigate the dose dependent nuclease activity of CP-Cas9 recombinant protein, the nuclease activity were measured in the same manner as in Example 8-1.


The mixture was mixed with 12.5, 25, 50, 100 or 200 ng of CP-Cas9 recombinant proteins and 50 ng of gRNA, and incubated with 300 ng of pUC19 plasmid for 1 hour at 37 V in 10 μl of reaction buffer.


As a positive control, the pUC19 plasmid was treated with restriction enzyme, NdeI.


As shown in FIG. 25, 200 ng of the CP-Cas9 recombinant protein has an excellent nuclease activity.


8-4. Nuclease Activity: Targeted Digestion

For the in vitro plasmid cleavage assay, SureGuide Cas9 Nuclease kit (Agilent, Calif., USA) was used. As a control, commercial Cas9 protein (Toolgen, Korea) was used.


250 ng of Cas9 proteins (Toolgen, Korea) or CP-Cas9 recombinant proteins were incubated with 100 ng of target DNA (Control DNA) and 50 ng of gRNA for 30 min at 30° C. in 20 μl of reaction buffer. The mixture was incubated at 65° C. for 15 minutes for inactivation. DNA cleavage products were separated by 1% agarose gel electrophoresis. When the target DNA was cleaved by Cas9, fragments of the target DNA were detected such as positive control.


As shown in FIG. 26, the CP-Cas9 recombinant protein has an excellent biological activity as nuclease compared to positive control.


EXAMPLE 9
Determination of Biological Activity of CP-Cas9 Recombinant Proteins in the in Reporter Cells

To investigate the biological activity of CP-Cas9 recombinant proteins at a cell level, HeLa cells transfected with the RFP plasmid were used as color reporter cells (FIG. 27). When the CP-Cas9 recombinant protein and gRNA are treated in the transfected HeLa cells, red fluorescence protein (RFP) is unexpressed.


As a control, commercial Cas9 protein (Toolgen, Korea) was used. gRNA was synthesized using a GeneArt™ Precision gRNA Synthesis kit (Thermo Fisher Scientific, Calif., USA). The gRNA was recognized part or all of the mRFP sequence of the plasmid as a target sequence. The target sequence has a nucleotide sequence selected from the group consisting of 5′-CTCCTCCGAGGACGTCATCA-3′, 5′-CAAGGAGTTCATGCGCTTCA-3′, 5′- CGCTTCAAGGTGCGCATGGA-3′. Preferably, the target sequence has 5′- CGCTTCAAGGTGCGCATGGA-3′.


The color reporter cells were treated with Cas9 protein or CP-Cas9 recombinant protein and gRNA according to 3 protocols, the protocols are as follows.


9-1. Determination of Biological Activity of CP-Cas9 Recombinant Proteins in the in Reporter Cells: Protocol 1

HeLa cells transfected with the RFP plasmid were seeded in a 24-well plate (1×105/well). After a day, 120 ng of gRNA (5′-CGCTTCAAGGTGCGCATGGA-3′) and 500 ng of CP-Cas9 recombinant protein were mixed within Opti-MEM (Thermo Fisher), and the mixture incubated at 37° C. for 15 minutes. The mixture were mixed with Lipofectamin 3000 (Thermo Fisher Scientific, Calif., USA) and incubated at RT for 15 minutes. Then, the cells were treated with the mixture for 48 hours. The cells were observed red fluorescence protein (RFP) by fluorescence microscopy.


As shown in FIG. 28, the CP-Cas9 recombinant protein showed low level of RFP expression, compare to vehicle. RFP expression of CP-Cas9 recombinant protein were similar to that of Cas9. As a result, it was confirmed that the CP-Cas9 recombinant protein deletes the target sequence or gene in the nucleus by CRISPR/Cas system.


9-2. Determination of Biological Activity of CP-Cas9 Recombinant Proteins in the in Reporter Cells: Protocol 2

HeLa cells transfected with the RFP plasmid were seeded in a 6-well plate (0.7×105/well). After a day, 1 ug of gRNA (5′-CGCTTCAAGGTGCGCATGGA-3′) and 5 ul of Lipofectamin 3000 were mixed in Opti-MEM (Thermo Fisher), and the mixture incubated at RT for 15 minutes. The mixture were mixed within Serum free DMEM. Then, the cells were treated with the mixture. After 4 hours, the cells were washed twice with PBS, changed to DMEM with 10% FBS and incubated at 37 ° C.


Next day, CP-Cas9 recombinant proteins were mixed within serum free DMEM, and the cells were treated with the CP-Cas9 recombinant proteins. After 8 hours, the cells were washed twice with PBS, changed to DMEM with 10% FBS and incubated at 37° C. for 24 hours. The cells were observed red fluorescence protein (RFP) by fluorescence microscopy.


As shown in FIG. 29, the CP-Cas9 recombinant protein showed low level of RFP expression, compare to the Cas9 protein.


9-3. Determination of Biological Activity of CP-Cas9 Recombinant Proteins in the in Reporter Cells: Protocol 3

HeLa cells transfected with the RFP plasmid were seeded in a 24-well plate (1×105/well). After a day, 120 ng of gRNA (5′-CGCTTCAAGGTGCGCATGGA-3′) and 500 ng of CP-Cas9 recombinant protein were mixed within Opti-MEM (Thermo Fisher), and the mixture incubated at 37° C. for 15 minutes. The mixture were mixed within serum free DMEM. Then, the cells were treated with the mixture. The cells were observed red fluorescence protein (RFP) by fluorescence microscopy.


As shown in FIG. 30, the CP-Cas9 recombinant protein showed low level of RFP expression, compare to the Cas9 protein.


These results suggest that the CP-Cas9 recombinant protein fused aMTD is enhanced its tissue-permeability and therefore, aMTD is critical for systemic delivery of the protein in vitro.

Claims
  • 1. A Cell-Permeable (CP)-Cas9 recombinant protein, which comprises a Cas9 protein and at least one advanced macromolecule transduction domain (aMTD)(s) being composed of 9 to 13 amino acid sequences and having improved cell and/or tissue permeability, wherein the aMTD is fused to one end or both ends of the Cas9 protein and has the following features of: (a) being composed of 3 or more amino acids sequences selected from the group consisting of Ala, Val, Ile, Leu, and Pro;(b) having Proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and the last of its amino acid sequence; and(c) having an instability index of 40 to 60; an aliphatic index of 180 to 220; and a grand average of hydropathy (GRAVY) of 2.1 to 2.6, as measured by Protparam.
  • 2. The CP-Cas9 recombinant protein according to claim 1, wherein the CP-Cas9 recombinant protein further comprises one or more solubilization domain (SD)(s), and the aMTD(s), the Cas9 protein and the SD(s) are randomly fused to one another.
  • 3. The CP-Cas9 recombinant protein according to claim 1, wherein the aMTD(s) is composed of 12 amino acid sequences and represented by the following general formula:
  • 4. The CP-Cas9 recombinant protein according to claim 2, wherein the CP-Cas9 recombinant protein is represented by any one of the following structural formulae: A-B-C, A-C-B, B-A-C, B-C-A, C-A-B, C-B-A and A-C-B-Cwherein A is the aMTD(s) having improved cell and/or tissue permeability,B is the Cas9 protein, andC is the SD(s), and if the CP-Cas9 recombinant protein comprises two SDs, the two SDs can be same or different.
  • 5. The CP-Cas9 recombinant protein according to claim 1, wherein the Cas9 protein has an amino acid sequence of SEQ ID NO: 1221.
  • 6. The CP-Cas9 recombinant protein according to claim 5, wherein the Cas9 protein is encoded by a polynucleotide sequence of SEQ ID NO: 1222.
  • 7. The CP-Cas9 recombinant protein according to claim 1, wherein the at least one aMTD(s) has an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 1 to 240.
  • 8. The CP-Cas9 recombinant protein according to claim 7, wherein the at least one aMTD(s) is encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 241 to 480.
  • 9. The CP-Cas9 recombinant protein according to claim 2, wherein the one or more SD(s) has an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 1203 to 1209.
  • 10. The CP-Cas9 recombinant protein of claim 9, wherein the one or more SD(s) is encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 1210 to 1216.
  • 11. The CP-Cas9 recombinant protein according to claim 1, wherein the CP-Cas9 recombinant protein has a histidine-tag affinity domain additionally fused to one end thereof.
  • 12. The CP-Cas9 recombinant protein according to claim 11, wherein the histidine-tag affinity domain has an amino acid sequence of SEQ ID NO: 1217.
  • 13. The CP-Cas9 recombinant protein of claim 12, wherein the histidine-tag affinity domain is encoded by a polynucleotide sequence of SEQ ID NO: 1218.
  • 14. The CP-Cas9 recombinant protein according to claim 1, wherein the fusion is formed via a peptide bond or a chemical bond.
  • 15. The CP-Cas9 recombinant protein according to claim 1, wherein the CP-Cas9 recombinant protein is used for deletion, insertion or replacement of a target sequence or gene.
  • 16. A polynucleotide sequence encoding the CP-Cas9 recombinant protein of claim 1.
  • 17. A recombinant expression vector comprising the polynucleotide sequence of claim 16.
  • 18. A transformant transformed with the recombinant expression vector of claim 17.
  • 19. A preparing method of the CP-Cas9 recombinant protein comprising: preparing the recombinant expression vector of claim 17;preparing a transformant using the recombinant expression vector;culturing the transformant; andrecovering the recombinant protein expressed by the culturing.
  • 20. A method of inserting, replacing or deleting of a target sequence or gene in a subject comprising: identifying a subject in need of inserting, replacing or deleting a target sequence or gene; andadministering to the subject an effective amount of the CP-Cas9 recombinant protein of claim 1.
PCT Information
Filing Document Filing Date Country Kind
PCT/KR2017/010747 9/27/2017 WO 00
Provisional Applications (1)
Number Date Country
62400861 Sep 2016 US