Cell permeable Nm23 recombinant proteins, polynucleotides encoding the same, and anti-metastatic composition comprising the same

Information

  • Patent Grant
  • 8450279
  • Patent Number
    8,450,279
  • Date Filed
    Thursday, September 4, 2008
    15 years ago
  • Date Issued
    Tuesday, May 28, 2013
    11 years ago
Abstract
The present invention discloses cell permeable Nm23 recombinant proteins where a macromolecule transduction domain (MTD) is fused to a metastasis suppressor Nm23. Also disclosed are polynucleotides encoding the cell permeable Nm23 recombinant proteins, an expression vector containing the cell permeable Nm23 recombinant protein, and a pharmaceutical composition for preventing metastasis which contains the cell permeable Nm23 recombinant protein as an effective ingredient. The cell permeable Nm23 recombinant proteins of the present invention can induce KSR phosphorylation and inactivation and inhibit Ras-mediated MAPK cascade by efficiently introducing a metastasis suppressor Nm23 into a cell. Therefore, the cell permeable Nm23 recombinant proteins of the present invention can be effectively used as an anti-metastatic agent capable of preventing cancer metastasis by inhibiting the proliferation, differentiation and migration of cancer cells.
Description
TECHNICAL FIELD

The present invention relates to cell permeable Nm23 recombinant proteins in which a metastasis suppressor Nm23 is fused to a macromolecule transduction domain (MTD), polynucleotides encoding the same, expression vectors for producing the same, and anti-metastatic pharmaceutical compositions including the same as effective ingredients for inhibiting metastasis.


BACKGROUND ART

Nm23 gene has been reported to encode proteins involved in the development and differentiation of a normal tissue, and its expression is decreased in various metastatic cells. Nm23 proteins belong to a tumor metastasis suppressor and generally consist of 150 to 180 amino acids. Nm23 proteins contain a leucine zipper motif and exhibit nucleoside diphosphate kinase (NDPK) activity. Two human Nm23 homologues, Nm23-H1 and Nm23-H2, consist of 152 amino acids having molecular weights of 17,143 and 17,294, respectively. In particular, it has been found that Nm23-H1 plays an important role in tumor metastasis and other various cell mechanisms including cell proliferation, embryogenesis, differentiation and tumorigenesis.


The mechanisms by which Nm23 affects tumor development and metastasis have not yet been clearly investigated. NDPK transfers a phosphoryl group between nucleoside triphosphate and nucleoside diphosphate via a covalent phosphoenzyme intermediate. For such a phosphorylation, histidine 118 of each Nm23-H1 and Nm23-H2 is served as a target site. Apart from the NDPK-mediated histidine phosphorylation, serine autophosphorylation was observed in Nm23 (MacDonald N J et al., J. Biol. Chem. 268: 25780-25789, 1993). When melanoma cells of Nm23 transfected mice were compared with a control cell, there was a direct correlation between the in vivo phosphorylation level of Nm23 serine and the inhibition of tumor metastatic potential. Serine phosphorylation of mouse Nm23 is inhibited by cAMP in vivo, while by forskolin in vitro, which demonstrates that the phosphorylation is controlled by signal transduction pathways.


Initially, Nm23 expression was reported to closely correlate to mouse melanoma cell lines with poor metastatic potential. The relationship between Nm23 reduced expression and tumor metastasis has been regarded as direct evidence to support the fact that Nm23 functions as a tumor metastasis suppressor (Steeg, P. S., Breast Dis. 10: 47-50, 1998). Inducible overexpression of Nm23 exhibited remarkably reduced metastatic potential in a highly metastatic cancer cell line. The Nm23 gene cloned as a putative tumor metastasis suppressor gene exhibits serine/threonine specific phosphotransferase and histidine protein kinase activities, as well as NDPK activity. Further, the expression of Nm23 is reduced as hematopoietic stem cells (HSC) are differentiated, which suggests that Nm23 is an important factor for anti-differentiation in those cells (Gervasi, F. et al., Cell Growth Differ 7: 1689-95, 1996). It has been found that Nm23 exhibits a strong inhibitory effect on tumor metastasis upon temporary transfection in forced inducible gene expression and in vitro metastasis model systems of human tumors (Hirayama, R. et al., J. Natl. Cancer Inst. 83: 1249-50, 1991; Nakayama, T. et al., J. Natl. Cancer Inst. 84: 1349-54, 1992; Leone, A. et al., Oncogene 8: 2325-33, 1993; Leone, A. et al., Oncogene 8: 855-65, 1993). In contrast, Nm23 mutation, leading to the loss of NDPK activity, had no influence on the inhibitory function of Nm23 in breast cancer cells (MacDonald, N. J. et al., J. Biol. Chem. 271:25107-16, 1996).


The most authentic evidence for the fact that Nm23 is a metastasis suppressor is revealed when the Nm23 gene is transfected into tumor cell lines. In metastatically competent cells, the administration of high dose Nm23 showed reduced metastatic activity by 40 to 98% as compared with a control transfectant (Leone, A. et al, Cell 65: 25-35, 1991; Leone, A. et al., Oncogene 8:2325-33, 1993).


Recently, it has been reported that Nm23 interacts with a kinase suppressor of Ras (KSR) discovered in the Drosophilae (Drosophilar melanogaster) and nematode (Caenorhabditis elegans) systems (Morrison, D. K., J. Cell Sci. 114: 1609-12, 2001). KSR is a scaffold protein of a mitogen-activated protein kinase (MAPK) cascade (Burack, W. R. and Shaw, A. S., Curr. Opin. Cell Biol. 12: 211-6, 2000; Pawson, T. and Scott, J. D., Science 278: 2075-80, 1997). Such scaffold protein is necessary to enhance the rate of phosphorylation and contribute to the specificity and stabilization of the phosphorylation pathway. Once the MAPK signal transduction pathway is activated by active Ras, KSR is forcedly dephosphorylated, and then, serve as a scaffold for the activation of MAPK cascade. During this process, Nm23 phosphorylates KSR serine 392, which is a binding site for another associated protein of KSR. If the serine 392 is mutated, Nm23 phosphorylates KSR serine 434. The metastatic inhibitory activity of Nm23 has been clearly demonstrated by the fact that metastatic potential is inhibited in various tumor cells transfected with Nm23 gene (Yoshida, T. et al., J Gastroenterol. 35: 768-74, 2000). In cells activated by the stimulation of the MAPK cascade, the interaction between Nm23 and KSR induces KSR phosphorylation in vitro in a complicated manner via a histidine-dependent pathway (Hartsough, M. T. et al., J. Biol. Chem. 277: 32389-99, 2002). Further, the in vivo association of KSR and Nm23 inhibits the phenotypic effect of active Ras which activates the MAPK cascade.


Accordingly, the administration of high dose Nm23 protein may phosphorylate and inactivate KSR in vivo, leading to the inhibition of Ras-mediated MAPK cascade. The present inventors have therefore believed that the inhibition of MAPK signal transduction pathway mediated by re-phosphorylation of KSR may inhibit cell proliferation, differentiation and migration of cancer cells and exhibit anti-metastatic effect in various human cancers, and endeavored to develop new anti-metastatic agents by using the Nm23 protein.


Meanwhile, small molecules derived from synthetic compounds or natural compounds can be transported into the cells, whereas macromolecules, such as proteins, peptides, and nucleic acids, cannot. It is widely understood that macromolecules larger than 500 kDa are incapable of penetrating the plasma membrane, i.e., the lipid bilayer structure, of live cells. To overcome this problem, a macromolecule intracellular transduction technology (MITT) was developed (Jo et al., Nat. Biotech. 19: 929-33, 2001), which allows the delivery of therapeutically effective macromolecules into cells, making the development of new drugs using peptides, proteins and genetic materials possible. According to this method, if a target macromolecule is fused to a hydrophobic macromolecule transduction domain (MTD) and other cellular delivery regulators, synthesized, expressed, and purified in the form of a recombinant protein, it can penetrate the plasma membrane lipid bilayer of the cells, be accurately delivered to a target site, and then, effectively exhibit its therapeutic effect. Such MTDs facilitate the transport of many impermeable materials which are fused to peptides, proteins, DNA, RNA, synthetic compounds, and the like into the cells.


Accordingly, the inventors of the present invention have developed a method of mediating the transport of a metastasis suppressor Nm23 into the cells, where cell permeable Nm23 recombinant proteins are engineered by fusing a MTD to the metastasis suppressor Nm23. Such cell permeable Nm23 recombinant proteins have been found to efficiently mediate the transport of a metastasis suppressor Nm23 into the cells in vivo as well as in vitro and can be used as anti-metastatic agents for inhibiting metastasis occurring in various human cancers.


DISCLOSURE
Technical Problem

Accordingly, the objective of the present invention is to provide cell permeable Nm23 recombinant proteins as an anti-metastatic agent which is effective for preventing metastasis in various kinds of human cancers by inhibiting proliferation, differentiation and migration of cancer cells.


Technical Solution

One aspect of the present invention relates to cell permeable Nm23 recombinant proteins capable of mediating the transport of a metastasis suppressor Nm23 into a cell by fusing a macromolecule transduction domain (MTD) having cell permeability to the metastasis suppressor protein.


Another aspect of the present invention relates to polynucleotides encoding the above cell permeable Nm23 recombinant proteins.


The present invention also relates to expression vectors containing the above polynucleotides, and transformants transformed with the above expression vectors.


Another aspect of the present invention relates to a method of producing cell permeable Nm23 recombinant proteins involving culturing the above transformants.


Another aspect of the present invention relates to a pharmaceutical composition including the above cell permeable Nm23 recombinant proteins as an effective ingredient for inhibiting metastasis.





DESCRIPTION OF DRAWINGS


FIG. 1 is a schematic diagram illustrating the structures of cell permeable Nm23 recombinant proteins being fused to one of a kFGF4-derived MTD, JO-76 MTD and JO-77 MTD and constructed in the full-length forms according to the present invention.



FIG. 2
a is a photograph of an agarose gel electrophoresis analysis showing PCR-amplified DNA fragments encoding cell permeable Nm23 recombinant proteins being fused to a kFGF4-derived MTD and constructed in the full-length forms according to the present invention.



FIG. 2
b is a photograph of an agarose gel electrophoresis analysis showing PCR-amplified DNA fragments encoding cell permeable Nm23 recombinant proteins being fused to each of JO-76 and JO-77 MTDs and constructed in the full-length forms according to the present invention.



FIG. 3
a is a schematic diagram illustrating the subcloning of a PCR product encoding a cell permeable Nm23 recombinant protein into the pGEM-T Easy vector according to the present invention.



FIGS. 3
b and 3c are photographs of an agarose gel electrophoresis analysis showing the PCR products encoding the cell permeable Nm23 recombinant proteins from FIGS. 2a and 2b subcloned in the pGEM-T Easy vector according to the present invention, respectively.



FIG. 4
a is a schematic diagram illustrating the cloning of a recombinant DNA fragment encoding a cell permeable Nm23 recombinant protein into the pET 28(+) vector according to the present invention.



FIGS. 4
b and 4c are photographs of an agarose gel electrophoresis analysis showing the recombinant DNA fragments encoding the cell permeable Nm23 recombinant proteins subcloned in the pET 28(+) vector according to the present invention.



FIG. 5 is a photograph of a SDS-PAGE analysis showing the inducible expression of cell permeable Nm23 recombinant proteins according to the present invention in various kinds of host cells.



FIG. 6 is a photograph of a SDS-PAGE analysis showing the purification of cell permeable Nm23 recombinant proteins expressed from the transformants where the expression vector according to the present invention is transformed into.



FIGS. 7
a and 7b are graphs illustrating the results of flow cytometry analysis of cell permeabilities of cell permeable Nm23 recombinant proteins according to the present invention.



FIGS. 8
a to 8c are confocal laser scanning microscopy photographs visualizing the cell permeabilities of cell permeable Nm23 recombinant proteins according to the present invention in mouse fibroblasts.



FIGS. 9
a and 9b are photographs of a Western blot analysis showing the inhibitory effect of cell permeable Nm23 recombinant proteins according to the present invention on MAPK signal transduction.



FIGS. 10
a and 10b are photographs of an invasion analysis showing the inhibitory effect of cell permeable Nm23 recombinant proteins according to the present invention on metastasis.



FIG. 11 is a photograph of a wound migration assay showing the inhibitory effect of cell permeable Nm23 recombinant proteins according to the present invention on metastasis.



FIG. 12
a is a photograph illustrating the inhibitory effect on metastasis in a mouse lung tissue extracted from a mouse administered with the cell permeable Nm23 recombinant protein according to the present invention.



FIG. 12
b is a photograph of immunohistochemical staining showing the expression of a metastatic marker, vimentin, in a mouse lung tissue extracted from a mouse administered with the cell permeable Nm23 recombinant protein according to the present invention.



FIG. 13 is a photograph of a terminus deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) analysis showing the apoptosis-inducing effect in a mouse lung tissue extracted from a mouse administered with the cell permeable Nm23 recombinant protein according to the present invention.



FIG. 14 is a photograph of a microarray analysis showing differential gene expression in a mouse lung tissue extracted from a mouse administered with the cell permeable Nm23 recombinant protein according to the present invention.





BEST MODE FOR CARRYING OUT THE INVENTION

The present invention provides cell permeable Nm23 recombinant proteins (CP-Nm23) capable of mediating the transport of a metastasis suppressor Nm23 into a cell in which the metastasis suppressor Nm23 is fused to a macromolecule transduction domain and, thereby, imparted with cell permeability; and polynucleotides encoding each of the cell permeable Nm23 recombinant proteins.


The present invention is characterized in that a metastasis suppressor Nm23 which is a macromolecule incapable of being introduced into a cell is fused to a specific macromolecule transduction domain (hereinafter, “MTD”) peptide so as to provide cell permeability, and thus, can be effectively transported into a cell. The MTD peptide may be fused to the N-terminus, the C-terminus, or both termini of the metastasis suppressor Nm23.


The present invention relates to cell permeable Nm23 recombinant proteins that are engineered by fusing a metastasis suppressor Nm23 to one of three MTD domains capable of mediating the transport of a macromolecule into a cell.


The term “cell permeable Nm23 recombinant protein” as used herein refers to a covalent bond complex bearing a MTD and a metastasis suppressor protein Nm23, where they are functionally linked by genetic fusion or chemical coupling. Here, the term “genetic fusion” refers to a co-linear, covalent linkage of two or more proteins or fragments thereof via their individual peptide backbones, through genetic expression of a polynucleotide molecule encoding those proteins.


Nm23 is a metastasis suppressor protein which inhibits the proliferation, differentiation and migration of cancer cells and induces apoptosis by controlling the MAPK signal transduction cascade which is mediated by KSR phosphorylation. Nm23 has an amino acid sequence represented by SEQ ID NO: 2, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 1. Nm23 functions as an important target protein in signal transduction cascades including KSR and Ras-mediated MAPK.


It has been reported that Nm23 is an endogeneous protein and exhibits NDP (nucleotide diphosphate)-kinase enzyme activity (Biggs et al., Cell 63, 933-940, 1990). Nm23 has also been found to be a transcription factor and cell differentiation inhibitor (I factor) (Postel et al., Science 261, 478-480, 1993; Okabe-Kado et al., Biochim. Biophys. Acta. 1267, 101-106, 1995).


In humans, eight nm23 isotypes (nm23-H1, nm23-H2, DR-nm23, nm23-H4, nm23-H5, nm23-H6, nm23-H7, and nm23-H8) have been identified to date, all of which are implicated in the regulation of metastasis (Rosengard et al., Nature 342, 177-180, 1989; Charpin C. et al., Int. J. Cancer 74, 416-420, 1997). In certain embodiments, cell permeable recombinant proteins for Nm23-H1 have been constructed, but are not limited thereto.


For the MTD capable of being fused to the metastasis suppressor Nm23, cell permeable peptides having an amino acid sequence selected from the group consisting of SEQ ID NOS: 4, 6, 8, and 37 to 227 may be used. The MTD having one of the amino acid sequences represented by SEQ ID NOS: 4, 6, 8 and 37 to 227 is a cell permeable polypeptide which is capable of mediating the transport of a biologically active molecule, such as a polypeptide, a protein domain, or a full-length protein across the cell membrane. Suitable MTDs for the present invention include a hydrophobic region showing cell membrane targeting activity by forming a helix structure at a signal peptide which is composed of an N-terminal domain, a hydrophobic domain and a C-terminal domain containing a secreted protein cleavage site. These MTDs can directly penetrate the cell membrane without causing any cell damage, transport a target protein into a cell, and thus, allow the target protein to exhibit its desired function.


The MTDs having the amino acid sequences represented by SEQ ID NOS: 4, 6, 8, and 37 to 227 and capable of being fused to a metastasis suppressor Nm23 according to the present invention are summarized in the following Tables 1a to 11.












TABLE 1a





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-01
CAC04038 putative NLP/P60-family
Ala Val Val Val Cys Ala Ile
37



secreted protein [Streptomyces
Val Leu Ala Ala Pro





coelicolor A3(2)]








JO-02
NP_057021 phosphatidylinositol
Pro Leu Ala Leu Leu Val Leu
38



glycan, class T precursor
Leu Leu Leu Gly Pro




[Homo sapiens]







JO-03
NP_072171 chorionic
Leu Leu Leu Ala Phe Ala Leu
39



somatomammotropin hormone 2
Leu Cys Leu Pro




isoform 3 [Homo sapiens]







JO-04
NP_932156 nudix-type motif 9
Leu Leu Gly Ala Leu Ala Ala
40



isoform a [Homo sapiens]
Val Leu Leu Ala Leu Ala






JO-05
NP_057327 NAD(P)H: quinone
Pro Val Leu Leu Ala Leu Gly
41



oxidoreductase type 3, polypeptide A2
Val Gly Leu Val Leu Leu Gly




[Homo sapiens]
Leu Ala Val






JO-06
CAD55300 putative secreted protein.
Ala Ala Ala Ala Val Leu Leu
42



[Streptomyces coelicolor A3(2)]
Ala Ala






JO-07
NP_629514 secreted protein
Ile Val Val Ala Val Val Val Ile
43



[Streptomyces coelicolor A3(2)]







JO-08
CAB57190 putative secreted chitin
Ala Val Leu Ala Pro Val Val
44



binding protein [Streptomyces
Ala Val





coelicolor A3(2)]








JO-09
CAB51015 putative secreted protein
Leu Ala Val Cys Gly Leu Pro
45



[Streptomyces coelicolor A3(2)]
Val Val Ala Leu Leu Ala






JO-10
NP_625021 glycosyl hydrolase (secreted
Leu Gly Gly Ala Val Val Ala Ala
46



protein) [Streptomyces coelicolor A3(2)]
Pro Val Ala Ala Ala Val Ala Pro






JO-11
NP_630686 secreted protein
Leu Leu Leu Val Leu Ala Val
47



[Streptomyces coelicolor A3(2)]
Leu Leu Ala Val Leu Pro






JO-12
NP_057329 dehydrogenase/reductase
Leu Leu Ile Leu Leu Leu Leu
48



(SDR family) member 8 [Homo sapiens]
Pro Leu Leu Ile Val






JO-13
NP_639877 putative secreted protein
Leu Ala Ala Ala Ala Leu Ala
49



[Streptomyces coelicolor A3(2)]
Val Leu Pro Leu






JO-14
NP_699201 protease inhibitor 16
Phe Leu Met Leu Leu Leu Pro
50



precursor [Homo sapiens]
Leu Leu Leu Leu Leu Val Ala






JO-15
NP_639871 putative secreted protein
Ala Ala Ala Ala Ala Ala Leu
51



[Streptomyces coelicolor A3(2)]
Gly Leu Ala Ala Ala Val Pro Ala






JO-16
CAB85250 putative secreted protein
Leu Leu Leu Ala Ala Leu Leu
52



[Neisseria meningitidis Z2491]
Leu Ile Ala Phe Ala Ala Val



















TABLE 1b





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-17
NP_626397 small secreted hydrophilic
Ala Leu Ala Ala Val Val Leu Ile
53



protein [Streptomyces coelicolor A3(2)]
Pro Leu Gly Ile Ala Ala






JO-18
CAB38593 putative secreted protein
Ala Ala Leu Ala Leu Gly Val Ala
54



[Streptomyces coelicolor A3(2)]
Ala Ala Pro Ala Ala Ala Pro Ala






JO-19
CAB57190 putative secreted chitin
Ala Ala Leu Ile Gly Ala Val Leu
55



binding protein 
Ala Pro Val Val Ala Val




[Streptomyces coelicolor A3(2)]







JO-20
NP_626007 secreted cellulose-binding
Ala Ala Gly Ile Ala Val Ala Ile
56



protein [Streptomyces coelicolor A3(2)]
Ala Ala Ile Val Pro Leu Ala






JO-21
NP_625632 secreted protein
Ile Ala Val Ala Ile Ala Ala Ile
57



[Streptomyces coelicolor A3(2)]
Val Pro Leu Ala






JO-22
CAC31790 putative secreted protein
Val Ala Met Ala Ala Ala Ala Val
58



[Mycobacterium leprae]
Leu Ala Ala Pro Ala Leu Ala






JO-23
NP_630266 secreted Protein
Leu Ala Val Leu Val Leu Leu Val
59



[StrePtomyces coelicolor A3(2)]
Leu Leu Pro






JO-24
NP_630165 secreted Protein
Val Val Ala Val Leu Ala Pro Val
60



[StrePtomyces coelicolor A3(2)]
Leu






JO-25
NC_003888 secreted Protein
Ala Ala Leu Leu Leu Pro Leu Leu
61



[StrePtomyces coelicolor A3(2)]
Leu Leu Leu Pro






JO-26
NP_627363 secreted Protein
Pro Ala Ala Val Ala Ala Leu Leu
62



[StrePtomyces coelicolor A3(2)]
Val Ile






JO-27
NP_631288 secreted Protein
Leu Leu Ile Ala Ala Leu Leu Pro
63



[StrePtomyces coelicolor A3(2)]







JO-28
NP_630325 secreted Protein
Ala Ala Val Val Leu Leu Pro Leu
64



[StrePtomyces coelicolor A3(2)]
Ala Ala Ala Pro






JO-29
NP_631289 secreted Protein
Ala Ala Ala Ala Ala Ala Leu Leu
65



[StrePtomyces coelicolor A3(2)]
Val Pro






JO-30
CAB51015 Putative secreted Protein
Leu Pro Val Val Ala Leu Leu Ala
66



[StrePtomyces coelicolor A3(2)]







JO-31
NP_629515 chitinase C (secreted Protein)
Ala Ala Ala Leu Ala Ala Pro Leu
67



[StrePtomyces coelicolor A3(2)]
Ala Leu Pro






JO-32
NP_940995 C1q and tumor necrosis
Leu Leu Leu Ala Leu Leu Leu Ala
68



factor related Protein 1 isoform 1
Ala




[Homo saPiens]







JO-33
NP_854150 POSSIBLE CONSERVED SECRETED
Ala Val Ala Val Val Ala Leu Leu
69



PROTEIN [Mycobacterium bovis AF2122/97]



















TABLE 1c





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-34
NP_630361 Probable secreted Protein
Leu Leu Leu Ile Ile Val Leu Leu
70



[StrePtomyces coelicolor A3(2)]
Ile Val Pro






JO-35
P39790 Extracellular metalloProtease
Leu Ala Leu Ala Ala Ala Val Val
71



Precursor
Pro






JO-36
CAA19252 Putative liPoProtein
Pro Ala Ala Leu Ala Leu Leu Leu
72



[StrePtomyces coelicolor A3(2)]
Val Ala






JO-37
P_625685 large secreted Protein
Ile Val Ala Leu Leu Leu Val Pro
73



[StrePtomyces coelicolor A3(2)]
Leu Val Leu Ala Ile Ala Ala Val





Leu






JO-38
NP_625685 large secreted Protein
Ile Val Ala Leu Leu Leu Val Pro
74



[StrePtomyces coelicolor A3(2)]







JO-39
NP_625685 large secreted Protein
Pro Leu Val Leu Ala Ile Ala Ala
75



[StrePtomyces coelicolor A3(2)]
Val Leu






JO-40
NP_808800 golgi PhosPhoProtein 2
Pro Leu Val Leu Ala Ala Leu Val
76



[Homo sapiens]
Ala






JO-41
NP_626993 selected Protein
Ala Ala Ala Leu Leu Ala Val Ala
77



[StrePtomyces coelicolor A3(2)]







JO-42
NP_004863 thymic dendritic cell-
Pro Leu Leu Leu Leu Ala Leu Ala
78



derived factor 1 [Homo saPiens]







JO-43
NP_631398 secreted Protein
Ala Leu Ala Leu Val Val Ala
79



[StrePtomyces coelicolor A3(2)]







JO-44
NP_627373 Penicillin-binding Protein
Val Ala Ala Val Val Val Ala Ala
80



(secreted Protein)





[StrePtomyces coelicolor A3(2)]







JO-45
NP_056226 sulfatase modifying
Pro Leu Leu Pro Leu Leu Leu Leu
81



factor 2 [Homo sapiens]
Val






JO-46
NP_854998 Conserved hypothelial secreted
Val Val Leu Val Val Val Leu Pro
82



protein [Mycobacterium bovis AF2122/97]
Leu Ala Val Leu Ala






JO-47
NP_627512 secreted Protein
Ala Ala Ala Val Pro Val Leu Val
83



[StrePtomyces coelicolor A3(2)]
Ala Ala






JO-48
NP_110448 phospholipase A2, group
Pro Ala Leu Leu Leu Leu Leu Leu
84



XIIA [Homo sapiens]
Ala Ala Val Val






JO-49
NP_003245 tissue inhibitor of metallo-
Pro Leu Ala Ile Leu Leu Leu Leu 
85



proteinase 1 precursor [Homo sapiens]
Leu Ile Ala Pro






JO-50
NP_002978 small inducible cytokine A17
Pro Leu Leu Ala Leu Val Leu Leu
86



precursor [Homo sapiens]
Leu Ala Leu Ile Ala






JO-51
NP_001012495 stromal cell derived factor
Val Val Ala Val Leu Ala Leu Val
87



1 isoform gamma precursor [Mus musculus]
Leu Ala Ala Leu



















TABLE 1d





MTD
Origin
Amino acid sequence
SEQ ID NO


















J0-52
NP 775628 ficolin 3 isoform 2
Pro Leu Leu Leu Leu Leu Pro Ala
88



precursor [Homo sapiens]
Leu






JO-53
NP 624483 secreted protein
Leu Ala Ala Val Ala Ala Leu Ala
89



[Streptomyces coelicolor A3(2)]
Val Val Val Pro






JO-54
NP_997465 HERV-FRD provirus
Leu Leu Leu Leu Val Leu Ile Leu
90



ancestral Env polyprotein [Homo sapiens]
Pro Leu Ala Ala






JO-55
NP_854234 posible conserved secreted
Leu Ala Val Val Val Val Ala Ala
91



protein [Mycobacterium bovis AF2122/97]
Val






JO-56
P23284 Peptidyl-prolyl cis-trans
Val Leu Leu Ala Ala Ala Leu Ile
92



isomerase B precursor (PPIase)
Ala




(Rotamase) (Cyclophilin B)







JO-57
CAD05047 hypothetical secreted protein
Leu Ile Ala Leu Leu Ala Ala Pro
93



[Salmonella enterica subsp.
Leu Ala





Enterica serovar Typhi]








JO-58
P05067 Amyloid beta A4 protein
Leu Ala Leu Leu Leu Leu Ala Ala
94



precursor (APP) (ABPP) (Alzheimer





disease amyloid protein)







JO-59
NP_004878 small inducible cytokine
Leu Leu Ala Ala Ala Leu Leu Leu
95



B14 precursor [Homo sapiens]
Leu Leu Leu Ala






JO-60
NP_626589 secreted protein
Val Ile Ile Ala Leu Ile Val Ile
96



[Streptomyces coelicolor A3(2)]
Val Ala






JO-61
NP_626589 secreted protein
Val Val Leu Val Val Ala Ala Val
97



[Streptomyces coelicolor A3(2)]
Leu Ala Leu






JO-62
NP_856548 SOLUBLE SECRETED ANTIGEN MPB53
Val Ala Val Ala Ile Ala Val Val
98



[Mycobacterium bovis AF2122/97]
Leu






JO-63
NP_629854 secreted protein
Pro Leu Ile Val Val Val Ala Ala
99



[Streptomyces coelicolor A3(2)]
Ala Val Val Ala Val






JO-64
AAB59058 lambda receptor protein
Pro Leu Ala Val Ala Val Ala Ala
100



[Escherichia coli]
Val Ala Ala






JO-65
NP_825185 NLP/P60-family secreted
Ala Ala Ile Ala Leu Val Ala Val
101



protein
Val Leu




[Streptomyces avermitilis MA-4680]







JO-66
NP_626568 secreted protein
Ala Ala Ala Leu Ala Ala Ile Ala
102



[Streptomyces coelicolor A3(2)]
Val Ile






JO-67
NP_626568 secreted protein
Ala Ala Ala Pro Ala Val Ala Ala
103



[Streptomyces coelicolor A3(2)]



















TABLE 1e





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-68
NP_625639 secreted protein
Leu Leu Leu Ala Ala Leu Pro
104



[Streptomyces coelicolor A3(2)]







JO-69
CAC32053 putative secreted protein
Ala Leu Leu Ala Val Val Ala Ala
105



[Mycobacterium leprae]







JO-70
NP_630954 secreted protein
Ala Val Val Val Val Leu Pro Ile
106



[Streptomyces coelicolor A3(2)]
Leu Leu






JO-71
P97300 Neuroplastin precursor
Ala Leu Ala Leu Leu Leu Leu Val
107



(Stromal cell-derived receptor 1)
Pro




(SDR-1)







JO-72
AAA41949 Rat parotid gland acidic
Leu Val Val Leu Leu Ala Ala Leu
108



proline-rich protein mRNA, S
Leu Val Leu




complete CD







JO-73
AAA17887 Drosophila melanogaster
Pro Val Leu Leu Leu Leu Ala Pro
109



spatzle (spz) gene







JO-74
NP_627867 conserved secreted protein
Ala Leu Ala Val Val Ala Ala Pro
110



[Streptomyces coelicolor A3(2)]







JO-75
NP_631283 secreted protein
Val Ile Val Ala Leu Leu Ala Val
111



[Streptomyces coelicolor A3(2)]







JO-76
NP_003231 endometrial bleeding 
Ala Leu Val Leu Pro Leu Ala Pro
  6



associated factor preproprotein





[Homo sapiens]







JO-77
CAB76313 putative secreted protein
Ala Val Ala Leu Leu Ile Leu Ala
  8



[Streptomyces coelicolor A3(2)]
Val






JO-78
P07198 Xenopsin precursor [Contains:
Val Leu Leu Ala Val Ile Pro
112



Xenopsin precursor fragment (XPF);





Xenopsin]







JO-79
NP_631293 secreted protein
Leu Ile Val Ala Ala Val Val Val
113



[Streptomyces coelicolor A3(2)]
Val Ala Val Leu Ile






JO-80
NP_626373 secreted protein
Ala Val Val Val Ala Ala Pro
114



[Streptomyces coelicolor A3(2)]







JO-81
NP_624952 secreted cellulose-binding
Leu Ala Ala Val Leu Leu Leu Ile
115



protein [Streptomyces coelicolor A3(2)]
Pro






JO-82
NP_009104 protease, serin, 23 
Leu Leu Leu Leu Leu Leu Ala Val
116



precursor [Homo sapiens]
Val Pro






JO-83
AAK63068 phytotoxic protein PcF
Ala Val Ala Leu Val Ala Val Val
117



precursor [Phytophthora cactorum]
Ala Val Ala






JO-84
NC_003903 Streptomyces coelicolor
Leu Val Ala Ala Leu Leu Ala Val
118



A3(2) plasmid SCP1, complete sequence
Leu



















TABLE 1f





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-85
NP_629842 peptide transport system
Leu Leu Ala Ala Ala Ala Ala
119



secreted peptide binding protein
Leu Leu Leu Ala




[Streptomyces coelicolor A3(2)]







JO-86
NP_854067 Posible secreted protein
Leu Ala Val Leu Ala Ala Ala Pro
120



[Mycobacterium bovis AF2122/97]







JO-87
NP_627802 secreted protein
Val Val Val Leu Leu Val Leu Leu
121



[Streptomyces coelicolor A3(2)]
Ala Leu Val Val Val






JO-88
NP_627802 secreted protein
Val Val Ile Ala Val Val Pro
122



[Streptomyces coelicolor A3(2)]







JO-89
NP_624483 secreted protein
Leu Ala Ala Val Ala Ala Leu Ala
123



[Streptomyces coelicolor A3(2)]
Val Val






JO-90
NP_627802 secreted protein
Val Leu Leu Val Leu Leu Ala Leu
124



[Streptomyces coelicolor A3(2)]
Val






JO-91
NP_625203 secreted protein
Pro Val Leu Val Pro Ala Val Pro
125



[Streptomyces coelicolor A3(2)]







JO-92
NP_630960 secreted protein
Pro Ala Leu Ala Leu Ala Leu Ala
126



[Streptomyces coelicolor A3(2)]







JO-93
NP_630670 secreted protein
Ala Ala Ala Ala Pro Ala Leu Ala
127



[Streptomyces coelicolor A3(2)]







JO-94
NP_630493 secreted protein
Ile Val Leu Pro Val Leu Ala Ala
128



[Streptomyces coelicolor A3(2)]
Pro






JO-95
CAC29994 putative secreted
Leu Val Leu Leu Leu Leu Pro Leu
129



protein [Mycobacterium leprae]
Leu Ile






JO-96
NP_624483 secreted protein
Leu Ala Ala Val Ala Pro Ala Leu
130



[Streptomyces coelicolor A3(2)]
Ala Val Val






JO-97
NP_037375 secretogranin III
Ile Leu Val Leu Val Leu Pro Ile
131



[Homo sapiens]







JO-98
NP_009199 V-set and
Ile Leu Leu Pro Leu Leu Leu Leu
132



immunoglobulin domain containing
Pro




4 [Homo sapiens]







JO-99
NP_733650 secreted hydrolase
Ile Ala Pro Ala Val Val Ala Ala
133



[Streptomyces coelicolor A3(2)]
Leu Pro






JO-100
NP_057540 transmembrane protein 9
Leu Leu Leu Val Ala Val Val Pro
134



[Homo sapiens]
Leu Leu Val Pro






JO-76
CAI74362 hypothetical protein
Leu Ile Leu Leu Leu Leu Pro Ile
135



[Theileria annulata]
Ile






JO-102
NP-630671 secreted protein
Ala Val Leu Ala Ala Pro Ala Val
136



[Streptomyces coelicolor A3(2)]
Leu Val






JO-77
NP_065695 TMEM9 domain family,
Leu Ala Leu Pro Val Leu Leu Leu
137



member B [Homo sapiens]
Ala






JO-104
P06908 Pulmonary surfactant-
Leu Ala Leu Ala Leu Leu Leu
138



associated protein A precursor





(SP-A) (PSP-A) (PSAP)





















TABLE 1g





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-105
NP_639721 putative secreted protein
Val Ala Val Pro Leu Leu Val Val
139



[Streptomyces coelicolor A3(2)]
Ala






JO-106
NP_854954 CONSERVED PROBABLE SECRETED
Ala Val Ala Val Ala Pro Val Ala
140



PROTEIN [Mycobacterium bovis AF2122/97]
Ala Ala Ala






JO-107
NP_627759 secreted protein
Ala Ala Ala Val Val Ala Ala Val
141



[Streptomyces coelicolor A3(2)]
Pro Ala Ala






JO-108
NP_003842 cellular repressor of
Ala Leu Leu Ala Ala Leu Leu Ala
142



E1A-stimulated genes [Homo sapiens]
Pro






JO-109
NP_003842 cellular repressor of
Leu Leu Ala Leu Leu Val Pro
143



E1A-stimulated genes [Homo sapiens]







JO-110
NP_003842 cellular repressor of
Ala Leu Leu Ala Ala Leu Leu Ala
144



E1A-stimulated genes [Homo sapiens]
Leu Leu Ala Leu Leu Val






JO-111
NP_000589 Homo sapiens insulin-like
Ala Ala Ala Leu Pro Leu Leu Val
145



growth factor binding protein 3 (IGFBP3)
Leu Leu Pro






JO-112
CAB594459 putative secreted protein
Ala Ala Ala Val Pro Ala Ala Leu
146



[Streptomyces coelicolor A3(2)]
Ala Pro






JO-113
NP_628917 secreted protein
Ala Ala Leu Ala Val Ala Ala Leu
147



[Streptomyces coelicolor A3(2)]
Ala Ala






JO-114
NP_624695 secreted protein
Ala Val Leu Ala Ala Ala Val Pro
148



[Streptomyces coelicolor A3(2)]







JO-115
NP_624695 secreted protein
Val Ala Ala Leu Pro Ala Pro Ala
149



[Streptomyces coelicolor A3(2)]







JO-116
NP_624791 secreted protein
Ala Leu Ala Leu Ala Val Pro Ala
150



[Streptomyces coelicolor A3(2)]
Val Leu Pro






JO-117
CAB45579 putative secreted protein
Ala Ala Leu Leu Pro Ala Ala Val
151



[Streptomyces coelicolor A3(2)]
Ala Val Pro






JO-118
NP_627066 secreted protein
Ala Val Val Val Ala Leu Ala Pro
152



[Streptomyces coelicolor A3(2)]







JO-119
NP_630174 secreted substrate-binding
Ala Ala Ala Val Ala Leu Pro Ala
153



protein [Streptomyces coelicolor A3(2)]
Ala Ala Ala Leu Leu Ala






JO-120
P06727 Apolipoprotein A-IV precursor
Ala Val Val Leu Pro Leu Ala Leu
154



(Apo-AIV) (ApoA-IV) Homo sapiens
Val Ala Val Ala Pro






JO-121
Q62087 Serum paraoxonase/lactonase 3.
Leu Val Ala Leu Pro Leu Leu Pro
155




Mus musculus








JO-122
NP_627123 probable secreted penicillin-
Val Val Val Pro Leu Leu Leu Ile
156



binding protein
Val Pro




[Streptomyces coelicolor A3(2)]





















TABLE 1h





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-123
CAC30224 putative secreted 
Leu Ala Val Val Leu Ala Val
157



hydrolase [Mycobacterium leprae]
Pro






JO-124
OZZQAM circumsporozoite protein
Leu Leu Ala Val Pro Ile Leu
158



precursor-Plasmodium cynomolgi
Leu Val Pro






JO-125
Q15166 Serum paraoxonase/lactonase
Leu Val Ala Leu Val Leu Leu
159



3 [Homo sapiens]
Pro






JO-126
NP_060220 all-trans-13,14-
Leu Val Leu Leu Leu Ala Val
160



dihydroretinol saturase [Homo sapiens]
Leu Leu Leu Ala Val Leu Pro






JO-127
AL627273 Salmonella enterica serovar
Leu Leu Ala Pro Val Val Ala
161




Typhi (Salmonella typhi) strain CT18

Leu Val Ile Leu Pro






JO-128
NP_625987 secreted protein
Val Leu Ala Val Leu Ala Val
162



[Streptomyces coelicolor A3(2)]
Pro Val Leu Leu Leu Pro






JO-129
CAB45474 putative secreted protein
Val Val Ile Ala Val Val Pro
163



[Streptomyces coelicolor A3(2)]
Val Val Val






JO-130
CAB45474 putative secreted protein
Leu Leu Val Leu Leu Ala Leu
164



[Streptomyces coelicolor A3(2)]
Val Val Val Pro






JO-131
CAB36605 putative secreted protein
Val Leu Leu Ala Leu Pro Val
165



[Streptomyces coelicolor A3(2)]
Val Ala Ala Pro






JO-132
NP_628377NLP/P60-family secreted
Ala Val Val Val Pro Ala Ile
166



protein [Streptomyces coelicolor A3(2)]
Val Leu Ala Ala Pro






JO-133
CAB59594 putative secreted protein
Ala Val Leu Val Pro Ala Ala
167



[Streptomyces coelicolor A3(2)]
Ala Leu Val Pro






JO-134
NP_624974 secreted protein
Val Val Ala Ala Leu Pro Leu
168



[Streptomyces coelicolor A3(2)]
Val Leu Pro






JO-135
NP_733682 secreted ATP/GTP binding
Ala Ala Val Ala Leu Pro Ala
169



protein [Streptomyces coelicolor A3(2)]
Ala Ala Pro






JO-136
P27169 Serum paraoxonase/arylesterase 1
Leu Ile Ala Leu Pro Leu Leu
170



(PON 1) (Serum aryldialkylphosphatase 1)
Pro




(A-esterase 1) Homo sapiens







JO-137
P52430 Serum paraoxonase/arylesterase 1
Leu Leu Ala Leu Pro Leu Val
171



(PON 1) (Serum aryldialkylphosphatase 1)
Leu Val Leu Ala Leu Pro




(A-esterase 1) Homo sapiens



















TABLE 1i





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-138
NP_626569 secreted protein
Ile Val Pro Leu Leu Leu Ala Ala
172



[Streptomyces coelicolor A3(2)]
Pro






JO-139
NP_940995 C1q and tumor necrosis
Leu Leu Leu Ala Pro Leu Leu Leu
173



factor related protein 1 isoform 1
Ala Pro




[Homo sapiens]







JO-140
NP_626174 large secreted protein
Leu Ala Ala Leu Pro Val Ala Ala
174



[Streptomyces coelicolor A3(2)]
Val Pro






JO-141
CAB83860 putative protein-export
Ala Leu Ala Val Ile Val Leu Val
175



integral membrane protein 
Leu Leu




[Neisseria meningitidis Z2491]







JO-142
NP_001009551 cornichon-like
Leu Ala Leu Leu Leu Pro Ala Ala
176



isoform 2 [Homo sapiens]
Leu Ile






JO-143
NP_626808 secreted protein
Ala Leu Leu Pro Leu Leu Ala Val
177



[Streptomyces coelicolor A3(2)]
Val Leu Pro






JO-144
NP_639798 putative secreted protein
Ala Ile Ala Val Pro Val Leu Ala
178



[Streptomyces coelicolor A3(2)]
Ala Pro






JO-145
NP_000492 Homo sapiens elastin
Ala Ala Ala Pro Val Leu Leu Leu
179



(supravalvular aortic stenosis)
Leu Leu






JO-146
NP_630680 secreted sugar binding
Ala Ala Ala Val Ala Val Leu Ala
180



protein [Streptomyces coelicolor A3(2)]
Leu Ala Pro






JO-147
CAB56129 putative secreted protein
Ala Ala Leu Ala Ala Leu Val Val
181



[Streptomyces coelicolor A3(2)]
Ala Ala Pro






JO-148
NP_625109 secreted solute-binding lipo-
Ala Ala Leu Ala Ala Val Pro Leu
182



protein [Streptomyces coelicolor A3(2)]
Ala Leu Ala Pro






JO-149
NP_733579 secreted sugar-binding
Ala Leu Ala Val Ala Ala Pro Ala
183



protein [Streptomyces coelicolor A3(2)]
Leu Ala Leu Leu Pro






JO-150
NP_630126 secreted chitinase
Ala Ala Leu Pro Ala Ala Ala Pro
184



(secreted protein)





[Streptomyces coelicolor A3(2)]







JO-151
NP_630126 secreted chitinase
Ala Ala Ala Pro Val Ala Ala Val
185



(secreted protein)
Pro




[Streptomyces coelicolor A3(2)]







JO-152
NP_872425 secretory protein
Leu Leu Ala Val Leu Leu Ala Leu
186



LOC348174 [Homo sapiens]
Leu Pro






JO-153
NP_630107 secreted protein
Val Leu Ala Leu Leu Val Ala Val
187



[Streptomyces coelicolor A3(2)]
Val Pro




















TABLE 1j





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-154
NP_733688 peptide-binding transport
Ala Leu Val Val Pro Ala Ala Val
188



protein [Streptomyces coelicolor A3(2)]
Pro






JO-155
NP_629904 secreted protein
Ala Val Val Leu Pro Leu Leu Leu
189



[Streptomyces coelicolor A3(2)]
Pro






JO-156
YP_177852 MCE-FAMILY PROTEIN MCE3A
Ala Val Ile Pro Val Ala Val Leu
190



[Mycobacterium tuberculosis H37Rv]
Val Pro






JO-157
CAA19627 putative secreted solute
Ala Ala Ala Val Pro Ala Ala Val
191



binding protein
Leu Ala Pro




[Streptomyces coelicolor A3(2)]







JO-158
NP_639884 putative large secreted
Val Ala Val Pro Val Val Leu Ala
192



protein [Streptomyces coelicolor A3(2)]
Ile Leu Pro






JO-159
P24327 Foldase protein prsA precursor
Ile Ala Ile Ala Ala Ile Pro Ala
193




Ile Leu Ala Leu






JO-160
CAB84808 putative membrane lipoprotein
Ala Leu Ile Ala Pro Ala Leu Ala
194



[Neisseria meningitidis Z2491]
Ala Pro






JO-161
NP_639883 putative large secreted
Ala Ala Ile Ala Leu Val Ala Pro
195



protein [Streptomyces coelicolor A3(2)]
Ala Leu






JO-162
NP_639883 putative large secreted
Leu Ala Pro Ala Val Ala Ala Ala
196



protein [Streptomyces coelicolor A3(2)]
Pro






JO-163
NP_627362 secreted protein
Val Ala Ile Ile Val Pro Ala Val
197



[Streptomyces coelicolor A3(2)]
Val Ala Ile Ala Leu Ile Ile






JO-164
NP_627362 secreted protein
Ala Val Val Ala Ile Ala Leu Ile
198



[Streptomyces coelicolor A3(2)]
Ile






JO-165
NP_624625 secreted protein
Leu Ala Ala Val Pro Ala Ala Ala
199



[Streptomyces coelicolor A3(2)]
Pro






JO-166
NP_624625 secreted protein
Ala Val Ala Ala Leu Pro Leu Ala
200



[Streptomyces coelicolor A3(2)]
Ala Pro






JO-167
NP_624625 secreted protein
Leu Ala Ala Pro Ala Ala Ala Ala
201



[Streptomyces coelicolor A3(2)]
Pro






JO-168
NP_626936 secreted protein
Leu Ala Ala Val Val Pro Val Ala
202



[Streptomyces coelicolor A3(2)]
Ala Ala Val Pro






JO-169
NP_626936 secreted protein
Val Ala Ala Pro Ala Ala Ala Ala
203



[Streptomyces coelicolor A3(2)]
Pro






JO-170
NP_626936 secreted protein
Ala Val Pro Val Pro Val Pro Leu
204



[Streptomyces coelicolor A3(2)]



















TABLE 1k





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-171
NP_085072 matrilin 2 isoform b
Leu Leu Ile Leu Pro Ile Val Leu
205



precursor [Homo sapiens]
Leu Pro






JO-172
CAB94057 putative secreted protein
Ala Leu Ala Leu Pro Ala Leu Ala
206



[Streptomyces coelicolor A3(2)]
Ile Ala Pro






JO-173
NP_624384 secreted protein
Ala Val Ile Pro Ile Leu Ala Val
207



[Streptomyces coelicolor A3(2)]
Pro






JO-174
NP_733505 large, multifunctional
Leu Ile Leu Leu Leu Pro Ala Val
208



secreted protein
Ala Leu Pro




[Streptomyces coelicolor A3(2)]







JO-175
CAB45630 putative secreted protein
Ile Val Leu Ala Pro Val Pro Ala
209



[Streptomyces coelicolor A3(2)]
Ala Ala






JO-176
NP_627887 secreted protein
Val Val Val Val Pro Val Leu Ala
210



[Streptomyces coelicolor A3(2)]
Ala Ala Ala






JO-177
P06832 Bacillolysin precursor
Leu Val Ala Val Ala Ala Pro
211





JO-178
NP_625998 secreted protein
Leu Val Leu Ala Ala Pro Ala Ala
212



[Streptomyces coelicolor A3(2)]
Leu Pro






JO-179
NP_625057 secreted protein
Leu Ile Ala Pro Ala Ala Ala Val
213



[Streptomyces coelicolor A3(2)]
Pro






JO-180
NP_443750 ADP-ribosyltransferase
Ala Leu Ala Ala Leu Pro Ile Ala
214



5 precursor [Homo sapiens]
Leu Pro






JO-181
CAB84257 putative secreted protein
Ala Val Leu Leu Leu Pro Ala Ala
215



[Neisseria meningitidis Z2491]
Ala






JO-182
P00634 Alkaline phosphatase
Ile Ala Leu Ala Leu Leu Pro Leu
216



precursor (APase)
Leu






JO-183
NP_000933 peptidylprolyl isomerase
Val Leu Leu Ala Ala Ala Leu Ile
217



B precursor [Homo sapiens]
Ala Pro






JO-184
CAB71258 putative secreted protein
Ala Pro Ala Val Leu Pro Pro Val
218



[Streptomyces coelicolor A3(2)]
Val Val Ile






JO-185
CAC31847 possible secreted protein
Val Val Gly Leu Leu Val Ala Ala
219



[Mycobacterium leprae]
Leu






JO-186
NP_626948 secreted protein
Ala Ala Ile Ala Ala Ala Ala Pro
220



[Streptomyces coelicolor A3(2)]
Leu Ala Ala






JO-187
NP_059120 cat eye syndrome critical
Leu Leu Leu Ala Val Ala Pro
221



region protein 1 isoform a 





precursor [Homo sapiens]







JO-188
NP_006519 tissue factor pathway
Leu Ile Leu Leu Leu Pro Leu Ala
222



inhibitor 2 [Homo sapiens]
Ala Leu




















TABLE 1l





MTD
Origin
Amino acid sequence
SEQ ID NO







JO-189
P97299 Secreted frizzled-relating
Ala Leu Leu Leu Leu Val Leu
223



protein 2 precursor (sFRP-2)
Ala




(Secreted apoptosis-relating protein 1)







JO-190
NP_071447 tubulointerstitial
Leu Leu Leu Leu Leu Leu Pro
224



nephritis antigen-like 1
Leu Ala






JO-191
NP_056322 epidermal growth factor-like
Leu Ala Leu Pro Leu Leu Leu
225



protein 6 precursor [Homo sapiens]
Pro






JO-192
NP_628035 secreted penicillin-binding
Leu Leu Val Leu Pro Leu Leu
226



protein [Streptomyces coelicolor A3(2)]
Ile






JO-193
NP_683880 cathepsin H isoform b
Leu Pro Leu Leu Pro Ala Ala
227



precursor [Homo sapiens]
Leu Val






kFGF4-
kaposi fibroblast growth factor 4,
Ala Ala Val Leu Leu Pro Val
228


derived
kFGF4
Leu Leu Ala Ala Pro



MTD









In some embodiments, the present invention may employ a kaposi fibroblast growth factor 4 (kFGF4)-derived MTD having the nucleotide sequence of SEQ ID NO: 3 and the amino acid sequence of SEQ ID NO: 4 (hereinafter, “MTD1”), a JO-76 MTD having the nucleotide sequence of SEQ ID NO: 5 and the amino acid sequence of SEQ ID NO: 6 which is a hypothetical protein derived from Theileria annulata (hereinafter, “MTD2”), and a JO-77 MTD having the nucleotide sequence of SEQ ID NO: 7 and the amino acid sequence of SEQ ID NO: 8 which belongs to member B of a human TMEM9 domain family (hereinafter, “MTD3”), as the MTD capable of mediating the transport of the metastasis suppressor Nm23 into a cell.


The cell permeable Nm23 recombinant proteins according to the present invention have a structure where one of the three MTDs (kFGF4-derived MTD: MTD1, JO-76: MTD2, JO-77: MTD3) is fused to one terminus or both termini of a metastasis suppressor protein Nm23, and a SV40 large T antigen-derived nuclear localization sequence (NLS)(nucleotide sequence of SEQ ID NO: 9, amino acid sequence of SEQ ID NO:10) and a histidine-tag (His-Tag) affinity domain for easy purification are fused to one terminus of the resulting construct.


In another embodiment, the present invention relates to the construction of eight full-length forms of a cell permeable Nm23 recombinant protein by using one of a kFGF4-derived MTD, a JO-76 MTD and a JO-77 MTD.


As used herein, the term “full-length form” refers to a construct including the entire amino acid sequence of the metastasis suppressor protein Nm23 which does not contain any deletion, addition, insertion or substitution of one or more amino acid residues in the amino acid sequence of SEQ ID NO: 2. However, it should be obvious to a skilled person in the art that Nm23 derivatives including various kinds of modifications through the deletion, addition, insertion or substitution of one or more amino acid residues in the amino acid sequence of SEQ ID NO: 2 that is made within the scope of not causing a deterioration of the Nm23 anti-metastatic effect can be used in the present invention.


Referring to FIG. 1, the full-length forms of the cell permeable Nm23 recombinant protein are as follows:


1) His-MTD1-Nm23 (HM1N), where a kFGF4-derived MTD is fused to the N-terminus of a full-length Nm23,


2) His-Nm23-MTD1 (HNM1), where a kFGF4-derived MTD is fused to the C-terminus of a full-length Nm23,


3) His-MTD1-Nm23-MTD1 (HM1NM1) where a kFGF4-derived MTD is fused to both termini of a full-length Nm23,


4) His-MTD2-Nm23 (HM2N), where a JO-76 MTD is fused to the N-terminus of a full-length Nm23,


5) His-Nm23-MTD2 (HNM2), where a JO-76 MTD is fused to the C-terminus of a full-length Nm23,


6) His-MTD3-Nm23 (HM3N), where a JO-77 MTD is fused to the N-terminus of a full-length Nm23,


7) His-Nm23-MTD3 (HNM3), where a JO-77 MTD is fused to the C-terminus of a full-length Nm23, and


8) His-MTD3-Nm23-MTD3 (HM3NM3), where a JO-77 MTD is fused to both termini of a full-length Nm23,


where a His-tag and a NLS derived from SV40 large T antigen are covalently coupled to the N-terminus of the above constructs.


As for the full-length forms of the cell permeable Nm23 recombinant protein constructed by using a kFGF4-derived MTD as described above, His-MTD1-Nm23 (HM1N) has an amino acid sequence represented by SEQ ID NO: 22, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 21; His-Nm23-MTD1 (HNM1) has an amino acid sequence represented by SEQ ID NO: 24, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 23; and His-MTD1-Nm23-MTD1 (HM1NM1) has an amino acid sequence represented by SEQ ID NO: 26, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 25.


As for the full-length forms of the cell permeable Nm23 recombinant protein constructed by using a JO-76 MTD as described above, His-MTD2-Nm23 (HM2N) has an amino acid sequence represented by SEQ ID NO: 28, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 27; and His-Nm23-MTD2 (HNM2) has an amino acid sequence represented by SEQ ID NO: 30, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 29.


As for the full-length forms of the cell permeable Nm23 recombinant protein constructed by using a JO-77 MTD as described above, His-MTD3-Nm23 (HM3N) has an amino acid sequence represented by SEQ ID NO: 32, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 31; His-Nm23-MTD3 (HNM3) has an amino acid sequence represented by SEQ ID NO: 34, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 33; and His-MTD3-Nm23-MTD3 (HM3NM3) has an amino acid sequence represented by SEQ ID NO: 36, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 35.


As a control for the cell permeable Nm23 recombinant proteins, His-Nm23 (HN), where a full-length Nm23 is fused only to a NLS derived from SV40 large T antigen and a histidine-tag (His-Tag) without any MTD, is constructed. The control protein has an amino acid sequence represented by SEQ ID NO: 20, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 19.


Further, the present invention provides an expression vector containing the polynucleotide encoding each of the cell permeable Nm23 recombinant proteins described above, and a transformant capable of producing each of the cell permeable Nm23 recombinant proteins at high levels, which is obtainable by transforming a host cell using the expression vector.


As used herein, the term “expression vector” is a vector capable of expressing a target protein or a target RNA in a suitable host cell. The nucleotide sequence of the present invention may be present in a vector in which the nucleotide sequence is operably linked to regulatory sequences capable of providing for the expression of the nucleotide sequence by a suitable host cell.


Within an expression vector, the term “operably linked” is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence. 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 suitable for the present invention may include plasmid vectors, cosmid vectors, bacteriophage vectors, viral vectors and the like, but are not limited thereto. The expression vectors for use in the present invention 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 nucleotide sequence that enables the vector to replicate in the host cell in question.


The expression vector constructed according to the present invention may be exemplified by pET28a(+)-HNM1 where the polynucleotide encoding the recombinant protein HNM1 where a kFGF4-derived MTD is fused to the C-terminus of a full-length Nm23 is inserted into a cleavage site of NdeI restriction enzyme within the multiple cloning sites (MCS) of a pET-28a(+) vector.


In another embodiment, the polynucleotide of the present invention is cloned into a pET-28a(+) vector (NOVAGEN, USA) bearing a His-tag sequence so as to fuse six histidine residues to the N-terminus of the cell permeable Nm23 recombinant protein to allow easy purification.


Accordingly, the cell permeable Nm23 recombinant protein expressed in the above expression vector has a structure where one of a kFGF4-derived MTD, a JO-76 MTD and a JO-77 MTD is fused to the full-length or truncated Nm23, and a His-tag and NLS are linked to the N-terminus thereof.


The present invention further provides a transformant capable of producing each of the cell permeable Nm23 recombinant proteins at high levels which is obtainable by transforming a host cell using the expression vector. The host cell suitable for the present invention may be eukaryotic cells, such as E. coli. In one embodiment of the present invention, E. coli used as a host cell is transformed with the expression vector, for example, pET28a(+)-HNM1 containing the polynucleotide encoding the cell permeable recombinant protein HNM1 where a kFGF4-derived MTD is fused to the C-terminus of a full-length Nm23 according to the present invention so as to produce the cell permeable Nm23 recombinant protein at high levels. Methods for transforming bacterial cells are well known in the art, and include, but are not limited to, biochemical means such as transformation, transfection, conjugation, protoplast fusion, calcium phosphate-precipitation, and application of polycations such as diethylaminoethyl (DEAE) dextran, and mechanical means such as electroporation, direct microinjection, microprojectile bombardment, calcium phosphate (CaPO4) precipitation, calcium chloride (CaCl2) precipitation, PEG-mediated fusion and liposome-mediated method.


In some embodiments, the transformants obtained by transforming E. coli DH5α with the expression vector containing the cell permeable Nm23 recombinant protein HM3N where a JO-77 MTD is fused to the N-terminus of a full-length Nm23, and the expression vector containing the cell permeable Nm23 recombinant protein HNM3 where a JO-77 MTD is fused to the C-terminus thereof, respectively, were deposited under accession numbers KCTC-11380BP and KCTC-11381BP, respectively, with the Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 52, Oun-Dong, Yusong-Ku, Taejon 305-333, Republic of Korea. All deposits referred to herein were made on Aug. 28, 2008 in accordance with the Budapest Treaty, and all restrictions imposed by the depositor on the availability to the public of the deposited biological material will be irrevocably removed upon the granting of the patent.


The present invention provides a method of producing the cell permeable Nm23 recombinant proteins at high levels, which includes the step of culturing the above transformant.


The method of the present invention may be carried out by culturing the transformant in a suitable medium under suitable conditions for expressing a cell permeable Nm23 recombinant protein of the present invention in the expression vector introduced into the transformant. Methods for expressing a recombinant protein by culturing a transformant are well known in the art, and for example, may be carried out by inoculating a transformant in a suitable medium for growing the transformant, performing a subculture, transferring the same to a main culture medium, culturing under suitable conditions, for example, supplemented with a gene expression inducer, isopropyl-J3-D-thiogalactoside (IPTG) and, thereby, inducing the expression of a recombinant protein. After the culture is completed, it is possible to recover a “substantially pure” recombinant protein from the culture solution. The term “substantially pure” means that the recombinant protein and polynucleotide encoding the same of the present invention are essentially free of other substances with which they may be found in nature or in vivo systems to the extent practical and appropriate for their intended use.


A recombinant protein of the present invention obtained as above may be isolated from the inside or outside (e.g., medium) of host cells, and purified as a substantially pure homogeneous polypeptide. The method for polypeptide isolation and purification is not limited to any specific method. In fact, any standard method may be used. For instance, chromatography, filters, ultrafiltration, salting out, solvent precipitation, solvent extraction, distillation, immunoprecipitation, SDS-polyacrylamide gel electrophoresis, isoelectric point electrophoresis, dialysis, and recrystallization may be appropriately selected and combined to isolate and purify the polypeptide. As for chromatography, affinity chromatography, ion-exchange chromatography, hydrophobic chromatography, gel filtration chromatography, reverse phase chromatography, adsorption chromatography, etc., for example, may be used (Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1982; Sambrook et al., Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, 1989; Deutscher, M., Guide to Protein Purification Methods Enzymology vol. 182. Academic Press. Inc., San Diego, Calif., 1990).


Meanwhile, the recombinant protein expressed in the transformants according to the present invention can be classified into a soluble fraction and an insoluble fraction according to protein characteristics during the protein purification process. If the majority of the expressed recombinant proteins are present in the soluble fraction, the recombinant protein can be isolated and purified according to the method as described above. However, when the majority of the expressed recombinant proteins are present in the insoluble fraction, i.e., as inclusion bodies, the recombinant proteins are first solubilized by using polypeptide denaturing agents, e.g., urea, guanidine HCl, or detergents, and then, purified by performing a series of centrifugation, dialysis, electrophoresis and column chromatography. Since there is the risk of losing the recombinant protein's activity due to a structural modification caused by the polypeptide denaturing agent, the process of purifying the recombinant protein from the insoluble fraction requires desalting and refolding steps. That is, the desalting and refolding steps can be performed by dialysis and dilution with a solution that does not include a polypeptide denaturing agent or by centrifugation with a filter. Further, if a salt concentration of the solution used for the purification of a recombinant protein from a soluble fraction is relatively high, such desalting and refolding steps may be performed.


In some embodiments, it has been found that the cell permeable Nm23 recombinant protein of the present invention mostly exists in the insoluble fraction as an inclusion body. In order to purify the recombinant protein from the insoluble fraction, the insoluble fraction may be dissolved in a lysis buffer containing a non-ionic surfactant such as Triton X-100, subjected to ultrasonification, and then centrifuged to separate a precipitate. The separated precipitate may be dissolved in a buffer supplemented with a strong denaturing agent, such as urea, and centrifuged to separate the supernatant. The above separated supernatant is purified by means of a histidin-tagged protein purification kit and subjected to ultrafiltration, for example, by using an amicon filter for salt removal and protein refolding, thereby obtaining a purified recombinant protein of the present invention.


Further, the present invention provides an anti-metastatic pharmaceutical composition including the cell permeable Nm23 recombinant protein as an effective ingredient for preventing metastasis by inhibiting the proliferation, differentiation and migration of cancer cells and inducing apoptosis.


The metastasis suppressor Nm23, which functions as an important target protein for signal transduction cascades including KSR and Ras-mediated MAPK, can control the MAPK signal transduction cascade mediated by KSR phosphorylation, and thus, inhibit the proliferation, differentiation and migration of cancer cells and induce apoptosis. Therefore, the cell permeable Nm23 recombinant proteins of the present invention can be effectively used as an anti-metastatic agent capable of preventing and/or treating cancer metastasis.


The cell permeable Nm23 recombinant proteins of the present invention can activate cell signaling mechanisms involved in the activation of ATM and p53 that induce cell cycle arrest and apoptosis in response to DNA damage or oncogenic signals by efficiently introducing a metastasis suppressor protein Nm23 into a cell. Therefore, the cell permeable Nm23 recombinant proteins of the present invention can be effectively used as an anti-metastatic agent for treating various kinds of human cancers.


The pharmaceutical composition comprising the recombinant protein of the present invention as an effective ingredient may further include pharmaceutically acceptable carriers suitable for oral administration or parenteral administration. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art (Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Company, Easton, Pa., 1995). The carriers for oral administration may include lactose, starch, cellulose derivatives, magnesium stearate, stearic acid and the like. In case of oral administration, the recombinant protein of the present invention can be formulated in the form of chewable tablets, buccal tablets, troches, capsules, elixir, suspensions, syrup, wafers or combination thereof by mixing with the carriers. Further, the carriers for parenteral administration may include water, suitable oil, saline, aqueous glucose, glycol and the like, and may further include stabilizers and preservatives. The stabilizers suitable for the present invention may include antioxidants such as sodium bisulfite, sodium sulfite and ascorbic acid. Suitable preservatives may include benzalconium chloride, methly-paraben, propyl-paraben and chlorobutanol.


The pharmaceutical composition of the present invention may be formulated into various parenteral or oral administration forms. Representative examples of the parenteral formulation include those designed for administration by injection. For injection, the recombinant proteins of the present invention may be formulated in aqueous solutions, specifically in physiologically compatible buffers or physiological saline buffer. These injection formulations may be formulated by conventional methods using one or more dispersing agents, wetting agents and suspending agents. For oral administration, the proteins can be readily formulated by combining the proteins with pharmaceutically acceptable carriers well known in the art. Such carriers enable the proteins of the invention to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Such oral solid formulations may include suitable excipients such as diluents (e.g., lactose, dextrose, sucrose, mannitol, sorbitol cellulose and/or glycin) and lubricants (e.g., colloidal silica, talc, stearic acid, magnesium stearate, calcium stearate, and/or polyethylene glycol). The tablets may include binders, such as aluminum silicate, starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP), and disintegrating agents, such as cross-linked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. If desired, absorbents, coloring agents, flavoring agents and/or sweeteners may be added. The formulations can be prepared by mixing, granulating or coating according to conventional methods well-known in the art.


If necessary, the pharmaceutical compositions of the present invention may further include pharmaceutical additives, such as preservatives, antioxidants, emulsifiers, buffering agents and/or salts for regulating osmosis and other therapeutically effective materials, and can be formulated according to conventional methods known in the art.


In addition, the pharmaceutical composition of the present invention can be administered via oral routes or parenteral routes such as intravenously, subcutaneously, intranasally or intraperitoneally. The oral administration may include sublingual application. The parenteral administration may include drip infusion and injection such as subcutaneous injection, intramuscular injection, intravenous injection and introtumoral injection.


The total effective amount of the recombinant protein of the present invention can be administered to patients in a single dose or can be administered by a fractionated treatment protocol, in which multiple doses are administered over a more prolonged period of time. Although the amount of the recombinant protein or nucleic acid encoding the same in the pharmaceutical composition of the present invention may vary depending on the severity of diseases, the protein or the nucleic acid may be generally administered several times a day at an effective dose of 5 to 20 mg. However, a suitable dose of the recombinant protein in the pharmaceutical composition of the present invention may depend on many factors, such as age, body weight, health condition, sex, disease severity, diet and excretion of patients, as well as the route of administration and the number of treatments to be administered. In view of the above factors, any person skilled in the art may determine the effective dose of the recombinant protein as an anti-metastatic agent for preventing metastasis in various human cancers. The pharmaceutical composition of the present invention containing the recombinant protein has no special limitations on its formulation, administration route and/or administration mode insofar as it exhibits the effects of the present invention.


EXAMPLES

The following examples are provided to illustrate the embodiments of the present invention in more detail, but are by no means intended to limit its scope.


Example 1
Construction of Cell Permeable Nm23 Recombinant Proteins (CP-Nm23)

Eight full-length forms of a cell permeable Nm23 (CP-Nm23) recombinant protein were constructed by using one of a kFGF4-derived MTD (MTD1), a JO-76MTD (MTD3) and a JO-77 MTD (MTD2) as a macromolecule transduction domain.


Referring to FIG. 1, the full-length forms of CP-Nm23 recombinant constructs were as follows:


1) His-MTD1-Nm23 (HM1N), where a kFGF4-derived MTD is fused to the N-terminus of a full-length Nm23,


2) His-Nm23-MTD1 (HNM1), where a kFGF4-derived MTD is fused to the C-terminus of a full-length Nm23,


3) His-MTD1-Nm23-MTD1 (HM1NM1) where a kFGF4-derived MTD is fused to both termini of a full-length Nm23,


4) His-MTD2-Nm23 (HM2N), where a JO-76 MTD is fused to the N-terminus of a full-length Nm23,


5) His-Nm23-MTD2 (HNM2), where a JO-76 MTD is fused to the C-terminus of a full-length Nm23,


6) His-MTD3-Nm23 (HM3N), where a JO-77 MTD is fused to the N-terminus of a full-length Nm23,


7) His-Nm23-MTD3 (HNM3), where a JO-77 MTD is fused to the C-terminus of a full-length Nm23, and


8) His-MTD3-Nm23-MTD3 (HM3NM3), where a JO-77 MTD is fused to both termini of a full-length Nm23,


where a His-tag and a NLS derived from SV40 large T antigen are covalently coupled to the N-terminus of the above constructs.


In order to prepare the full-length CP-Nm23 recombinant constructs, polymerase chain reactions (PCRs) were carried out by using the oligonucleotides described in Table 1 below as a primer pair specific for each recombinant construct and a human Nm23 cDNA (SEQ ID NO: 1) as a template. The forward and reverse primers for amplifying HM1N have nucleotide sequences represented by SEQ ID NOS: 13 and 12, respectively; those for amplifying HNM1 have nucleotide sequences represented by SEQ ID NOS: 11 and 14, respectively; those for amplifying HM1NM1 have nucleotide sequences represented by SEQ ID NOS: 13 and 14, respectively; those for amplifying HM2N have nucleotide sequences represented by SEQ ID NOS: 15 and 12, respectively; those for amplifying HNM2 have nucleotide sequences represented by SEQ ID NOS: 11 and 16, respectively; those for amplifying HM3N have nucleotide sequences represented by SEQ ID NOS: 17 and 12, respectively; those for amplifying HNM3 have nucleotide sequences represented by SEQ ID NOS: 11 and 18, respectively; and those for amplifying HM3NM3 have nucleotide sequences represented by SEQ ID NOS: 17 and 18, respectively.


The oligonucleotides as a forward and reverse primer set specific for each recombinant protein are summarized in Table 2 below.











TABLE 2






SEQ



Primer
ID NO
Sequence







HN-5′
11
CCG CAT ATG GCC AAC TGT GAG CGT ACC


(30 nts)

TCC





HN-3′
12
CCG CAT ATG TCA TTC ATA GAT CCA GTT


(33 nts)

CTG AGC





HM1N-5′
13
CCG CAT ATG GCA GCC GTT CTT CTC CCT


(72 nts)

GTT CTT CTT GCC GCA CCC GCC AAC TGT




GAG CGT ACC TTC ATT GCG





HNM1-3′
14
CCG CAT ATG TCA GGG TGC GGC AAG AAG


(72 nts)

AAC AGG GAG AAG AAC GGC TGC TTC ATA




GAT CCA GTT CTG AGC ACA





HM2N-5′
15
CCG CAT ATG GCG CTG GTG CTG CCG CTG


(60 nts)

GCG CCG GCC AAC TGT GAG CGT ACC TTC




ATT GCG





HNM2-3′
16
CCG CAT ATG TCA CGG CGC CAG CGG CAG


(60 nts)

CAC CAG CGC TTC ATA GAT CCA GTT CTG




AGC ACA





HM3N-5′
17
CCG CAT ATG GCG GTG GCG CTG CTG ATT


(63 nts)

CTG GCG GTG GCC AAC TGT GAG CGT ACC




TTC ATT GCG





HNM3-3′
18
CCG CAT ATG TCA CAC CGC CAG AAT CAG


(63 nts)

CAG CGC CAC CGC TTC ATA GAT CCA GTT




CTG AGC ACA









The PCR was performed in a 50 μl reaction mixture containing 100 ng of human Nm23 cDNA as a template, 0.2 mM dNTP mixture (dGTP, dATP, dTTP, and dCTP, each at 2 mM), 0.6 μM of each primer, 5 of 10×Taq buffer, 1 μl of Taq polymerase (TAKARA, Japan). The PCR was performed for 25 cycles at 94° C. for 45 seconds, at 53° C. for 45 seconds and at 72° C. for 45 seconds after the initial denaturation of 94° C. for 2 minutes, followed by the final extension at 72° C. for 5 minutes. After the PCR was completed, the amplified PCR product was digested with restriction enzyme Ndel and loaded onto a 1.0% agarose gel and fractionated. As shown in FIGS. 2a and 2b, it was confirmed that the expected fragment for each recombinant construct fused to one of a kFGF4-derived MTD, a JO-76 MTD and a JO-77 MTD was successfully amplified.


The DNA band of expected size was excised from the gel, eluted, and purified by using a QIAquick™ Gel extraction kit (QIAGEN, USA). The eluted DNA was precipitated with ethanol and resuspended in distilled water for ligation. As shown in FIG. 3a, the PCR amplified DNA fragment containing the coding region was subcloned into a pGEM-T Easy vector (PROMEGA, Madison Wis. USA) with a T4 ligase according to the TA cloning method, and then, followed by transformation of E. coli DH5α competent cells with the pGEM-T Easy vector. The cells were plated onto LB plate media supplemented with 50 μg/ml of ampicillin and cultured at 37° C. for overnight. After the recombinant fragment-inserted pGEM-T Easy vector was isolated by treating with restriction enzyme Ndel 37° C. for 1 hour, it was subjected to a 0.8% agarose gel electrophoresis.


As shown in FIG. 3b, the DNA fragments of about 0.5 kb for the full-length forms and vector fragments of about 3 kb were detected, confirming that the insert DNA of the CP-Nm23 recombinant construct was appropriately subcloned into the pGEM-T Easy vector.


A pET-28(+)a vector (NOVAGEN, Madison, Wis.) bearing a histidine-tag and a T7 promoter was digested with a restriction enzyme Ndel ( ENZYNOMICS, Korea). The pGEM-T Easy vector fragments containing the CP-Nm23 recombinant fragment and pET-28(+)a vector fragment were purified by using a QIAquick™ Gel extraction kit. Each of the pGEM-T Easy vector fragments was cloned into the pre-treated pET-28a(+) with a T4 ligase at 16° C. for 12 hours, followed by transformation of E. coli DH5α competent cells with the resulting pET-28a(+) vector (FIG. 4a).


After the clones were treated with the restriction enzyme NdeI (Enzynomics, Korea) and subjected to 0.8% agarose gel electrophoresis, it was verified that DNA fragments of about 0.5 kb for the full-length forms and vector fragments of about 5 kb were detected, confirming the cloning of the insert DNA of CP-Nm23 recombinant construct into pET-28a(+) vector, as shown in FIG. 4b.


The successfully cloned expression vectors for expressing cell permeable Nm23 recombinant proteins were designated pET28a(+)-HM1N, pET28a(+)-HNM1, pET28a(+)-HM1NM1, pET28a(+)-HM2N, pET28a(+)-HNM2, pET28a(+)-HM3N, pET28a(+)-HNM3 and pET28a(+)-HM3NM3, respectively. Among them, the E. coli transformants DH5α/HM3Nm23 and DH5α/HNm23M3 obtained by transforming E. coli DH5α with the expression vectors pET28a(+)-HM3N and pET28a(+)-HNM3, respectively, were deposited on Aug. 28, 2008 in accordance with the Budapest Treaty under accession numbers KCTC-11380BP and KCTC-11381BP with the Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 52, Oun-Dong, Yusong-Ku, Taejon 305-333, Republic of Korea.


The results of sequencing analysis are as follows:


As for the full-length forms of the cell permeable Nm23 recombinant protein constructed by using a kFGF4-derived MTD, His-MTD1-Nm23 (HM1N) has an amino acid sequence represented by SEQ ID NO: 22, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 21; His-Nm23-MTD1 (HNM1) has an amino acid sequence represented by SEQ ID NO: 24, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 23; and His-MTD1-Nm23-MTD1 (HM1NM1) has an amino acid sequence represented by SEQ ID NO: 26, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 25.


As for the full-length forms of the cell permeable Nm23 recombinant protein constructed by using a JO-76 MTD, His-MTD2-Nm23 (HM2N) has an amino acid sequence represented by SEQ ID NO: 28, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 27; and His-Nm23-MTD2 (HNM2) has an amino acid sequence represented by SEQ ID NO: 30, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 29.


As for the full-length forms of the cell permeable Nm23 recombinant protein constructed by using a JO-77 MTD as described above, His-MTD3-Nm23 (HM3N) has an amino acid sequence represented by SEQ ID NO: 32, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 31; His-Nm23-MTD3 (HNM3) has an amino acid sequence represented by SEQ ID NO: 34, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 33; and His-MTD3-Nm23-MTD3 (HM3NM3) has an amino acid sequence represented by SEQ ID NO: 36, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 35.


As a control for the cell permeable Nm23 recombinant proteins, His-Nm23 (HN), where a full-length Nm23 is fused only to a nuclear localization sequence (NLS) derived from SV40 large T antigen and a histidine-tag (His-Tag) without any MTD, was constructed. The control protein has an amino acid sequence represented by SEQ ID NO: 20, while a polynucleotide encoding the same has a nucleotide sequence represented by SEQ ID NO: 19.


Example 2
Expression of Recombinant Proteins

<2-1>Selection of Optimal Bacterial Strains


To select the optimal bacterial strain for the expression of cell permeable Nm23 recombinant proteins prepared in Example 1 above, the following experiments were carried out in E. coli BL21(DE3), BL21-Gold(DE3), BL21-CodonPlus(DE3) and BL21-Gold(DE3) pLysS strains (STRATAGENE, USA), all of which contain the LacI promoter.


First, each of the expression vectors pET28a(+)-HM1N, pET28a(+)-HNM1, pET28a(+)-HM1NM1, and pHN (control) was transformed into E. coli BL21(DE3), BL21-Gold(DE3), BL21-CodonPlus(DE3) and BL21-Gold(DE3) pLysS strains, respectively, according to the heat shock method. Further, each of the expression vectors pET28a(+)-HM2N, pET28a(+)-HNM2, pET28a(+)-HM3N, pET28a(+)-HNM3 and pET28a(+)-HM3NM3 was transformed into E. coli BL21-Gold(DE3) strain, respectively, according to the heat shock method. After the transformation, the cells were cultured in an LB agar plate containing 50 μg/ml of kanamycin. Colonies formed on the plate were grown in 1 in of LB medium at 37° C. overnight, followed by culturing at 37° C. in 100 μl of LB medium with vigorous shaking until the optical density 600 (OD600) reached 0.5. IPTG (isopropyl-β-D-thiogalactoside) was then added thereto at a final concentration of 0.7 mM to induce the expression of the CP-Nm23 recombinant proteins. Protein induction was prolonged for 2 hours at 30° C. The E. coli culture solutions were harvested by centrifugation at 4° C., 7,000×g for 20 minutes, resuspended in a lysis buffer (100 mM NaH2PO4, 10 mM Tris-HCl, 8 M urea, pH 8.0), and subjected to ultrasonication on ice using a sonicator equipped with a probe. The cell lysates were centrifuged at 14,000×g for 15 minutes, so as to separate an insoluble fraction from a soluble fraction. The thus obtained soluble and insoluble fractions of CP-Nm23 recombinant proteins expressed in the E. coli strain with IPTG were loaded on a SDS-PAGE gel.


As shown in FIG. 5, although some cell permeable Nm23 recombinant proteins were expressed at a relatively low level in BL21-Gold (DE3), most cell permeable Nm23 recombinant proteins showed the highest expression level in the strain. According to these results, BL21-Gold (DE3) was selected as the optimal strain for the expression of the cell permeable Nm23 recombinant proteins according to the present invention.


<2-2>Expression of Recombinant Proteins


Each of the expression vectors pET28a(+)-HM1N, pET28a(+)-HNM1, pET28a(+)-HM1NM1, pET28a(+)-HM2N, pET28a(+)-HNM2, pET28a(+)-HM3N, pET28a(+)-HNM3 and pET28a(+)-HM3NM3 was transformed into E. coli BL21-Gold(DE3), selected as the optimal strain in section <2-1> of Example 2 above, followed by inducing their expression through the addition of 0.7 mM IPTG, according to the same method described in section <2-1> of Example 2. After that, soluble and insoluble fractions of CP-Nm23 recombinant proteins obtained therefrom were loaded on a SDS-PAGE gel.


As shown in FIG. 6, it was confirmed that the cell permeable Nm23 recombinant proteins (19 to 20 kDa) expressed in the host cell were mostly included in the insoluble fraction as an inclusion body, and their expression was significantly increased in the presence of IPTG.


Example 3
Purification and Refolding of Recombinant Proteins

<3-1>Purification of Recombinant Proteins


The inducible expression of cell permeable Nm23 recombinant proteins in an E. coli system leads to the formation of insoluble aggregates, which are known as inclusion bodies. To completely solubilize these inclusion bodies, all of the above expressed proteins were denatured by dissolving them in 8 M urea used as a strong denaturing agent.


First, the BL21 Gold(DE3) strains transformed with each of the expression vectors pET28a(+)-HM1N, pET28a(+)-HNM1, pET28a(+)-HM1NM1, pET28a(+)-HM2N, pET28a(+)-HNM2, pET28a(+)-HM3N, pET28a(+)-HNM3, pET28a(+)-HM3NM3 and pET28a(+)-HM (control) were cultured in 1 l of an LB medium as described in Example 2. Each culture solution was harvested by centrifugation, gently resuspended in 20 of a lysis buffer (HN and HNM1: 50 mM NaH2PO4, 300 mM NaCl, 10 mM imidazole, pH 8.0; other CP-Nm23: 100 mM NaH2PO4, 10 mM Tris-HCl, 8 M urea, pH 8.0) without forming bubbles, and subjected to ultrasonication on ice using a sonicator equipped with a microtip. The cells were intermittently sonicated for 30 seconds, followed by cooling for 10 seconds, while setting the power to 25% of the maximum power. The total sonication time was 5 minutes. The cell lysates were centrifuged at 4° C., 4,000×g for 20 minutes, so as to separate the supernatant and the cellular debris pellet. The supernatant was loaded onto a Ni-NTA agarose resin where nitrilotriacetic acid agarose was charged with nickel (Ni). The Ni-NTA agarose resin was equilibrated with the lysis buffer. The supernatant was allowed to absorb onto the resin by gently shaking (using a rotary shaker) at 4° C. for 8 hours or more. The resin absorbed with the inclusion bodies containing the recombinant protein was centrifuged at 4° C., 1,000×g for 5 minutes, to remove the reaction solution and washed with a washing buffer (HN and HNM1: 50 mM NaH2PO4, 300 mM NaCl, 10 mM imidazole, pH 6.3; other CP-Nm23: 100 mM NaH2PO4, 10 mM Tris-HCl, 8 M urea, pH 6.3) five times to remove nonspecific absorbed materials. After washing, the proteins absorbed to the resin were eluted with an elution buffer (HN and HNM1: 50 mM NaH2PO4, 300 mM NaCl, 10 mM imidazole, pH 4.5; other CP-Nm23: 100 mM NaH2PO4, 10 mM Tris-HCl, 8 M urea, pH 4.5) with stirring for 2 hours or more under acidic conditions of pH 4.5. The eluted proteins were analyzed with 12% SDS-PAGE gel electrophoresis, stained with Coomassie Brilliant Blue R by gently shaking, and destained with a destaining solution.


According to the results shown in FIG. 6, all of the cell permeable Nm23 recombinant proteins fused to a kFGF4-derived MTD, a JO-76 MTD and a JO-77 MTD, respectively, were detected as a single band corresponding to about 19 to 20 kDa, which confirms that the cell permeable Nm23 recombinant proteins of the present invention have been purified from the insoluble fraction.


<3-2>Refolding of Recombinant Proteins


Since the cell permeable Nm23 recombinant proteins of the present invention purified from the insoluble fraction as described in section <3-1> of Example 3 above were denatured by a strong denaturing agent, such as 8 M urea, the denatured proteins must be converted into an active form by a refolding process, as follows.


First, the purified recombinant proteins were subjected to a refolding process by dialyzing them against a refolding buffer (0.55 M guanidine HCl, 0.88 M L-arginine, 50 mM Tris-HCl, 150 mM NaCl, 1 mM EDTA, 100 mM NDSB, 1 mM glutathione oxidized, and 1 mM glutathione reduced) at 4° C. for 24 hours, thereby removing the denaturing agent. All of the refolded recombinant proteins were dialyzed against a cell culture medium DMEM (Dulbecco's Modified Eagle Medium) by using a dialysis bag (Snakeskin pleated, PIERCE) at 4° C. for 10 hours while stirring. The medium was replaced with fresh DMEM every 3 hours. The cell permeable Nm23 recombinant proteins of the present invention converted into their active form through the refolding process were used in the following experiments.


Example 4
Quantitative Cell Permeability Analysis of Nm23 Recombinant Proteins

In order to quantitatively determine the cell permeability of the cell permeable Nm23 recombinant proteins according to the present invention, the introduction of the proteins into the cell was analyzed by FACS (fluorescence-activated cell sorting) in an animal model, as follows.


The cell permeable Nm23 recombinant proteins refolded into their active form in section <3-2> of Example 3 above were labeled with FITC (fluorescein-5-isothiocyanate, MOLECULAR PROBE). The recombinant protein (2 to 20 mg) was mixed with 1 μl of FITC at a concentration of 333 mg/ml and reacted in a dark room at room temperature for 1 hour with gentle stirring. The reaction solution was subjected to a dialysis against DMEM at 4° C. for 2 days until the unreacted FITC was completely removed, thereby obtaining FITC-conjugated recombinant proteins. Thus obtained FITC-conjugated recombinant proteins were subjected to a Bradford protein assay to measure the protein concentration. As a result, each of the FITC-conjugated recombinant proteins was measured to have a concentration of about 0.7 μg/μl.


Meanwhile, RAW 264.7 cells derived from mouse macrophage were maintained in DMEM supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin (500 mg/ml) and incubated at 37° C. in a humidified atmosphere of 5% CO2 in air.


After the incubation, the cells were treated with 10 μM of each of the FITC-conjugated recombinant proteins (HM1N, HNM1, HM1NM1, HM3N, HNM3 and HM3NM3) prepared above, followed by further culturing them for 1 hour at 37° C. Subsequently, the cells were treated with trypsin/EDTA (T/E, INVITROGEN) to remove cell surface bound proteins, washed with cold PBS (phosphate buffered saline) three times, and then, subjected to flow cytometry analysis by using a CELLQUEST PRO software program of the FACS (fluorescence-activated cell sorting) Calibur system ( BECKTON-DICKINSON). The cell concentration of each sample was 1×104 cells/μl, and the analysis was performed twice or more. The cell permeability of the cell permeable Nm23 recombinant proteins according to the present invention was determined by comparing it to that of the control protein (HN) not fused to a MTD.



FIGS. 7
a and 7b show the results of the flow cytometry analysis where the gray filled curve represents cell only, the black curve represents FITC only, the blue curve represents the cell permeability of the control protein not fused to a MTD (HN), the red curve represents the cell permeability of the cell permeable recombinant proteins HM1N, HM3N, HNM3 and HM3NM3, the green curve represents the cell permeability of the cell permeable recombinant protein HNM1, and the orange curve represents the cell permeability of the cell permeable recombinant protein HM1NM1. Referring to the results shown in FIGS. 7a and 7b, it was found that all of the cell permeable Nm23 recombinant proteins showed significantly higher cell permeability than the control protein.


Example 5
Microscopic Cell Permeability Analysis of Nm23 Recombinant Proteins

To visualize the intracellular localization of human Nm23 proteins delivered into a cell, NIH 3T3 cells (Korean Cell Line Bank, Seoul, Republic of Korea) were treated with FITC-conjugated recombinant proteins (HM1N, HNM1, HM1NM1, HM2N, HNM2, HM3N, HNM3 and HM3NM3) and visualized by confocal laser scanning microscopy.


First, the NIH 3T3 cells were cultured in an 8-well chamber slide (LabTek, Nalgen Nunc) for 24 hours. The NIH3T3 cells were maintained in DMEM supplemented with 10% fetal bovine serum, 5% penicillin/streptomycin (500 mg/ml) in 5% CO2 at 37° C. After the cells were washed with PBS three times, the cells were treated with serum-free DMEM, serum-free DMEM containing FITC, and serum-free DMEM containing 10 μM of each of FITC-conjugated recombinant proteins, respectively, in 5% CO2 at 37° C. One hour later, the cells were fixed with 4% paraformaldehyde at room temperature for 20 minutes.


For a direct detection of FITC-conjugated recombinant proteins that were internalized, the cells were washed with PBS three times and counterstained with a nuclear fluorescent stain solution, propidium iodide (PI, SIGMA-ALDRICH). The cells were stained with PI at a concentration of 1 μg/ml for 5 minutes, followed by washing with PBS three times. In order to preserve the FITC fluorescence of the recombinant protein, the glass slide was fixed in 10 μl of a polyvinyl alcohol mounting medium containing DABCO (Fluca) for 15 minutes before the observation. The intracellular distribution of the fluorescence was determined at the middle of a single cell analyzed by a confocal laser scanning microscope using a normaski filter. The confocal laser scanning microscopy was employed for observing cell phormology, FITC fluorescence and PI fluorescence. FITC was excited at 488 nm and detected by means of a bandpass filter at 530 nm.


As shown in FIGS. 8a to 8c, it was observed that the cell permeable Nm23 recombinant proteins stained with FITC and PI were well distributed largely in the nucleus as compared with the cell only, FITC only and MTD-lacking control protein. The intracellular localization of the cell permeable Nm23 recombinant proteins fused to one of a kFGF4-derived MTD, a JO-76 MTD and a JO-77 MTD according to the present invention was consistent with the cell permeability of the same determined by flow cytometry above. From these results, it was confirmed that the cell permeable Nm23 recombinant proteins of the present invention exhibited high cell permeability.


Example 6
Inhibitory Effect of Cell Permeable Nm23 Recombinant Proteins on MAPK Signal Transduction

In order to confirm the in vivo function of the cell permeable Nm23 recombinant proteins according to the present invention, the biochemical functions of the recombinant proteins were examined on three types of cancer cell lines by Western blot analysis.


MDA-MB-435 and MDA-MB-231 cells, the highly metastatic human breast cancer cell lines used in this experiment, were purchased from Korean Cell Line Bank (Seoul, Republic of Korea). The cell lines were maintained in a RPMI 1640 medium (L-glutamine 300 mg/l, 25 mM HEPES and 25 mM NaHCO3) supplemented with 10% FBS and 1% penicillin/streptomycin in a 5% CO2 incubator at 37° C. CCL-185 cells, a human lung cancer cell line, were obtained from ATCC and maintained in a HamF-12K medium (2 mM L-glutamine, 1500 mg/f sodium bicarbonate) supplemented with 10% FBS and 1% penicillin/streptomycin in a 5% CO2 incubator at 37° C.


After 2 ml of the RPMI 1640 medium supplemented with FBS was added to each well of a 6-well plate, MDA-MB-435, MDA-MB-231, and CCL-185 cells were inoculated thereto at a concentration of 5×106 cells/ml. The well plate was incubated at 37° C. for 1 day so as to allow the cells to grow while adhering to the well plate. After removing the medium, the cells adhered to the well plate were washed with cold PBS. Subsequently, the cells were treated with 500 μl of each of the cell permeable Nm23 recombinant proteins and MTD-lacking Nm23 control protein (HIN) at a concentration of 10 μM, and reacted in a 5% CO2 incubator at 37° C. for 1 hour. The MDA-MB-435 cells were treated with each of HM1N, HNM1, HM1NM1, HM2N, HNM2, HM3N, HNM3 and HM3NM3 recombinant proteins, while MDA-MB-231 and CCL-185 cells were treated with each of HM3N, HNM3 and HM3NM3 recombinant proteins. After the reaction was completed, the cells were washed twice with PBS, and then, cultured in the presence of serum under the same conditions noted above for 2, 4, 6 and 8 hours, respectively.


After the cultivation was completed, the cells were resuspended in 200 μl of a lysis buffer (20 mM HEPES, pH 7.2, 1% Triton-X, 10% glycerol and proteinase inhibitor) and subjected to ultrasonication on ice for 30 minutes, to thereby obtain a cell lysate. The cell lysate was centrifuged at 12,000 rpm for 20 minutes at 4° C. to separate the supernatant. The thus obtained supernatant was subjected to a Bradford protein assay to quantitatively measure the protein concentration. The recombinant protein was resuspended in a SDS-PAGE loading buffer at a concentration of 25 μM to prepare a cell lysate sample. The thus prepared cell lysate sample was heated at 90° C. for 5 minutes, and then, stored at −80° C. until use.


For the Western blot analysis, p21 (21 kDa, CELL SIGNALING TECHNOLOGY), phospho-p53 (Ser15, 53 kDa, Cell Signaling), phospho-MEK (Ser217/221, 45 kDa, CELL SIGNALING TECHNOLOGY), and phospho-Erk (Thr202/Tyr204, 42/44 kDa, CELL SIGNALING TECHNOLOGY) were used as primary antibodies, and goat anti-mouse IgG-HRP ( SANTA CRUZ BIOTECHNOLOGY) and goat anti-rabbit IgG-HRP (SANTA CRUZ BIOTECHNOLOGY) were used as secondary antibodies. The cell lysate sample was applied to a 12% SDS-PAGE at 100 V for 2 hours and transferred onto a polyvinylidene fluoride (PDVF) membrane at 100 V for 90 minutes. In order to prevent the nonspecific interaction between the blotted proteins and unrelated antibodies, the PVDF membrane was blocked with 5% non-fat dry milk in TBS/T (10 Mm Tris-Cl, pH 8.0, 150 mM NaCl, 0.05% Tween 20) at room temperature for 1 hour. After removing the blocking buffer, the PVDF membrane was washed with TBS/T, followed by incubation with each of the primary antibodies (diluted with the freshly prepared blocking buffer at a ratio of 1:10000) for 1 hour at 4° C. After removing the primary antibody solution, the membrane was washed with TBS/T five times each for 5 minutes, and incubated with the secondary antibody (diluted with the freshly prepared blocking buffer at a ratio of 1:5000) for 1 hour at room temperature. After washing with TBS/T five times, the membrane was stained using an enhanced chemiluminescence (ECL) detection system (GE HEALTHCARE AMERSHAM UK) to visualize the antigen/antibody interaction.


As shown in FIGS. 9a and 9b, in the cells treated with the cell permeable Nm23 recombinant protein as compared with cells treated with the control protein, the phosphorylation of KSR serine 392, which is a scaffold protein of the MAPK cascade, was enhanced, while the phosphorylation of MEK (P-MEK) that induces the activation of tumor cell cycle was reduced. In particular, the HM3N and HNM3 recombinant proteins where a JO-77 MTD was fused to the N- and C-terminus, respectively, strongly inhibited the phosphorylation of ERK and MEK in all three types of human cancer cell lines.


Example 7
In Vitro Anti-Metastatic Effect of Cell Permeable Nm23 Recombinant Proteins

<7-1>Invasion assay


In order to examine whether tumor metastasis is inhibited by blocking cancer cell migration in cancer cells treated with the cell permeable Nm23 recombinant proteins according to the present invention, an invasion assay was carried out as follows.


First, a human breast cancer cell line, MDA-MB-435 cells, were cultured overnight in a RPMI 1640 medium supplemented with 10% FBS in the absence of growth factors. The next day, the cells were treated with trypsin and harvested, followed by suspension in the same RPMI 1640 medium. The cells were treated with each of the MTD-lacking Nm23 control protein (HN), and cell permeable Nm23 recombinant proteins (HM2N, HNM2, HM3N, HNM3 and HM3NM3) according to the present invention at a concentration of 10 μM at 37° C. for 1 hour. Meanwhile, the top part of a trans-well polycarbonate membrane filter (BD Falcon) having a pore size of 3 μm was coated with MATRIGEL (40 μg per each well; BD Biosciences). To the lower part of the chamber, a DMEM medium supplemented with 10% FBS was added as an adhesive substrate. The cells treated with the above protein were suspended in a DMEM medium supplemented with 0.1% FBS to prepare a cell suspension. The thus prepared cell suspension was inoculated on the trans-well membrane filter (1×105 cells per each well), and cultured in a 5% CO2 incubator at 37° C. for 20 to 24 hours. The filters were washed with PBS, and the non-invasive cells remaining on the surface of the upper part were removed by using a cotton swab. The invasive cells that passed through the Matrigel and migrated to the lower part of the filter were fixed with 4% paraformaldehyde for 5 to 10 minutes, and stained with 0.5% (w/v) hemacolor for 10 to 20 minutes. The number of cells migrated to the base surface of the membrane filter (violet color) was counted by observing with an optical microscope.


According to the results shown in FIGS. 10a and 10b, in case of the cells treated with most of the cell permeable Nm23 recombinant proteins, HM2N, HM3N and HNM3 in particular, as compared with the control protein (HN), the invasion of the cells was significantly reduced. From these results, it was found that the cell permeable Nm23 recombinant proteins according to the present invention can effectively inhibit metastatic potential of tumor cells in vivo.


<7-2>Wound migration assay


In order to examine whether the cell permeable Nm23 recombinant proteins according to the present invention can inhibit the migration of a breast cancer cell line, MDA-MB-435 cells, having high migration activity, a wound migration assay was carried out as follows.


MDA-MB-435 cells were cultured in a 60-mm culture dish until they formed a confluent monolayer covering the bottom thereof. After incubation, the cells were treated with each of the MTD-lacking Nm23 control protein (HN) and cell permeable Nm23 recombinant proteins (HM2N, HNM2, HM3N, HNM3 and HM3NM3) according to the present invention at a concentration of 10 μM at 37° C. for 1 hour. After the cells were washed with PBS, they were wounded with a sterile yellow tip, to thereby form a reference line that separated the confluent area from the bare area. To the cells was added a RPMI medium (3 mL) supplemented with 10% FBS, followed by culturing in a 5% CO2 incubator at 37° C. for 24 hours. The cells were washed with PBS, fixed with methanol for 1 minute, stained with Giemsa (Chameleon Chemical) for 5 minutes, and then, washed with distilled water. The migration was quantified by counting the number of cells that migrated from the wound edge into the bare area with an inverted light microscope at 40× magnification.


Referring to the results shown in FIG. 11, the migration of tumor cells was remarkably inhibited in the cells treated with the cell permeable Nm23 recombinant proteins, HM2N, HM3N and HNM3 in particular, as compared with the control protein, which is consistent with the results from the invasion assay above.


Example 8
In Vivo Anti-Metastatic Effect of Cell Permeable Nm23 Recombinant Proteins

In order to examine the in vivo inhibitory effect of the cell permeable Nm23 recombinant proteins on tumor metastasis which has already been confirmed in vitro, an immunohistochemical analysis was carried out as follows.


First, MDA-MB-435 cells, a highly metastatic human breast cancer cell line, were suspended in 0.1 of PBS at a concentration of 1×106 cells/ml and were injected to the outer tail vein of 5-week old MHC (major histocompatibility complex)-deficient Balb/c nu/nu mice. Twenty mice were subdivided into 4 groups of 5 mice each. Each of the cell permeable Nm23 recombinant protein (HM3N, 300 μg) to which a JO-77 MTD was fused, a vehicle (PBS, 300 μg) and MTD-lacking Nm23 control protein (HN, 300 μg), and an EGFP recombinant protein (HM3E) where a JO-77 MTD was fused to the N-terminus of EGFP was administered to the mice. Here, the MTD-fused EGFP recombinant protein was employed as a control to examine whether the JO-77 MTD being fused to Nm23 had an effect on Nm23 expression. Five weeks after MDA-MB-435 cells were injected to the mice, the proteins were administered daily to the mice of each group via intravenous injection for 21 days. After three mice were selected from each group and sacrificed, lung tissue samples were extracted therefrom. The other two mice remaining in each group had undergone further observation for 14 days after the administration was terminated, and then, lung tissue samples were extracted therefrom. The lung tissue samples were fixed with a Bouin fixation solution overnight for detecting metastatic colonies, washed with distilled water, and then embedded in paraffin to prepare a paraffin block. Thus prepared paraffin block was sliced with a microtome to have a thickness of 4 μm, where the slices were mounted on a glass slide and treated with xylene for 5 minutes three times to remove paraffin. The glass slide was subjected to immunohistochemical staining with vimentin as a metastatic marker.


For the immunohistochemical staining, anti-vimentin antibody (ABCAM) was employed as a primary antibody, and goat anti-mouse IgG-HRP (Santa Cruz Biotechnology) was used as a secondary antibody. In order to prevent the nonspecific interaction between blotted proteins and irrelevant antibodies, the glass slide was blocked with 5% non-fat dry milk in TBS/T (10 mM Tris-Cl, pH 8.0, 150 mM NaCl, 0.05% Tween 20) at room temperature for 1 hour with stirring. After removing the blocking buffer, the glass slide was washed with TBS/T three times, followed by incubating with the anti-vimentin antibody as a primary antibody (diluted with PBS at a ratio of 1:200) for 1 hour at 4° C. After the removing the primary antibody solution, the glass slide was washed with TBS/T five times each for 5 minutes, and incubated with the goat anti-mouse IgG-HRP as a secondary antibody (diluted with PBS at a ratio of 1:200) for 1 hour at room temperature. After washing with TBS/T (0.025% Triton-X 100) twice, the glass slide was stained with a DAB substrate to detect vimentin.



FIG. 12
a shows the results of optically observing the lung tissue extracted from the mouse after the cell permeable Nm23 recombinant protein according to the present invention was administered for 21 days, and the same was extracted from the mouse where the administration of the cell permeable Nm23 recombinant protein was terminated for 14 days. As shown in FIG. 12a, in the lung tissues of mice treated with the vehicle, control protein (HN), and MTD-fused EGFP recombinant protein (HM3E), tumor growth was remarkably increased in spite of the treatment with the proteins for 21 days. Further, the tumor size was not reduced after the subsequent non-treatment period of 2 weeks and newly formed tumors were found in other peripheral tissues, showing that metastasis occurred. However, in the lung tissue of the mouse treated with the cell permeable Nm23 recombinant protein (HM3N) according to the present invention, there was no tumor formation not only during the 3-week period of protein treatment, but also during the subsequent 2-week period of the non-treatment, suggesting that the tumor formation and metastasis are effectively inhibited by the cell permeable Nm23 recombinant protein.



FIG. 12
b depicts the results of immunohistochemical staining showing the expression of a metastatic marker, vimentin, in the lung tissue extracted from the mouse after the cell permeable Nm23 recombinant protein according to the present invention was administered for 21 days (Day 21), and the lung tissue extracted from the mouse where the administration of the cell permeable Nm23 recombinant protein was terminated for 14 days (Day 35). As shown in FIG. 12b, vimentin was detected in the lung tissue of the mice treated with the vehicle, control protein (HN), and MTD-fused EGFP recombinant protein (HM3E) both on Day 21 and on Day 35, while vimentin was not detected in the lung tissue of the mouse treated with the cell permeable Nm23 recombinant protein (HM3N) according to the present invention neither on Day 21 nor on Day 35. From these results, it was confirmed that the cell permeable Nm23 recombinant protein according to the present invention can effectively inhibit tumor metastasis in vivo.


Example 9
In Vivo Apoptosis-Inducing Effect after the Administration of Cell Permeable Nm23 Recombinant Proteins

In order to examine the effect of inducing apoptosis in tumor tissues after the administration of the cell permeable Nm23 recombinant proteins, a TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling) assay was performed by using the same mouse model as described in Example 8. The TUNEL assay was carried out by using an in situ cell death detection kit (TMR red, ROCHE).


In particular, each of the cell permeable Nm23 recombinant protein (HM3N), vehicle and HN as a control, and MTD-fused EGFE recombinant protein (HM3E) was daily administered to the mice subdivided into four groups via intravenous injection for 21 days according to the same method as described in Example 8. After three mice were selected from each group and sacrificed, lung tissue samples were extracted therefrom. The other two mice remaining in each group had undergone further observation for 14 days after the administration was terminated, and then, lung tissue samples were extracted therefrom. The lung tissue samples were embedded in paraffin to prepare a paraffin block. Thus prepared paraffin block was sectioned with a microtome to have a thickness of 5 μm and mounted on a glass slide. The glass slide was treated with xylene for 5 minutes three times, to thereby remove paraffin. It was then successively treated with 100% ethanol twice for 5 minutes, and 90%, 80% and 70% ethanol each for 3 minutes so as to dehydrate the lung tissue, followed by incubation in PBS for 5 minutes. The glass slide was treated with 0.1% Trition® X-100 dissolved in a 0.1% sodium citrate solution for 8 minutes, and washed with PBS twice for 2 minutes. After a drop of TUNEL reaction buffer (50 μl, ROCHE, USA) was added to the glass slide, the glass slide was incubated in a humidified incubator at 37° C. for 1 hour, washed with PBS three times, and then, observed with a fluorescence microscope.


Referring to the results shown in FIG. 13, there was no significant histological change in the mouse lung tissue treated with the vehicle, control protein (FIN) and MTD-fused EGFP recombinant protein, while in the mouse lung tissue treated with the cell permeable Nm23 recombinant protein (HM3N), a region stained in red representing the characteristic of apoptosis was observed, confirming the effect of inducing apoptosis of the cell permeable Nm23 recombinant protein according to the present invention. Further, it was also observed that in the mouse lung tissue treated with the cell permeable Nm23 recombinant protein according to the present invention, apoptosis was still induced in cancer cells 14 days after the administration was terminated.


Example 10
Comparison of Protein Expression Pattern after the Administration of Cell Permeable Nm23 Recombinant Proteins

In order to examine the change in protein expression pattern in the tumor tissue treated with the cell permeable Nm23 recombinant protein according to the present invention, a microarray assay was performed as follows.


In particular, each of the cell permeable Nm23 recombinant protein (HM3N), vehicle and HN (control) was administered to the mice subdivided into three groups via intravenous injection for 21 days, and then left alone for 14 days after the administration was terminated, according to the same method as described in Example 9 above. Fourteen days after the administration was terminated, lung tissue samples were extracted from the mouse of each group and freezed with liquid nitrogen. Total RNA was isolated from the lung tissue by using a TRIZOL reagent (INVITROGEN) according to the manufacturer's instruction, and treated with an RNase-free DNase (LIFE TECHNOLOGIES, Inc.), to thereby completely remove the remaining genomic DNA.


The thus isolated RNA was subjected to synthesis and hybridization of a target cRNA probe by using a Low RNA Input Linear Amplification kit (Agilent Technology) according to the manufacturer's instruction. In brief, 1 μg of total RNA was mixed with a T7 promoter specific primer and reacted at 65° C. for 10 minutes. A cDNA master mix was prepared by mixing a first strand buffer (5×), 0.1 M DTT, 10 mM dNTP mix, RNase-Out and MMLV-RT (reverse transcriptase), and added to the reaction mixture. The resulting mixture was reacted at 40° C. for 2 hours, followed by reacting at 65° C. for 15 minutes, to thereby terminate the reverse transcription and dsDNA synthesis. A transcription master mix was prepared by mixing a transcription buffer (4×), 0.1 M DTT, NTP mix, 50% PEG, RNase-Out, inorganic pyrophosphatase, T7-RNA polymerase and cyanine (3/5-CTP) according to the manufacturer's instruction. The thus prepared transcription master mix was added to the dsDNA reaction mixture and reacted at 40° C. for 2 hours so as to perform dsDNA transcription. The thus amplified and labeled cRNA was purified with a cRNA Cleanup Module (AGILENT TECHNOLOGY) according to the manufacturer's instruction. The labeled target cRNA was quantified by using a ND-1000 spectrophotometer (NanoDrop Technologies, Inc.). After the labeling efficiency was examined, cRNA was mixed with a blocking agent (10×) and a fragmentation buffer (25×), and reacted at 60° C. for 30 minutes so as to carry out the fragmentation of cRNA. The fragmented cRNA was resuspended in a hybridization buffer (2×) and directly dropped on a Whole Human Genome Oligo Microarray (44K). The microarray was subjected to hybridization in a hybridization oven (Agilent Technology) at 65° C. for 17 hours, followed by washing according to the manufacturer's instruction (AGILENT Technology).


The hybridization pattern was read by using a DNA microarray scanner ( AGILENT Technology) and quantified by using a Feature Extraction Software ( AGILENT Technology). Data normalization and selection of fold-changed genes were carried out by using a Gene Spring GX 7.3 soft wear (AGILENT Technology). The average of the normalized ratio was calculated by dividing a normalized signal channel strength by a normalized control channel strength. Functional annotation for a gene was conducted by using a Gene Spring GX 7.3 software (AGILENT Technology) according to the Gene Ontology™ Consortium.


The results of the microarray analysis are summarized in FIG. 14 and Tables 3 to 9, where Table 3 shows the expression pattern of apoptosis-relating genes, Table 4 shows that of cell adhesion-relating genes, Table 5 shows that of cell cycle regulation-relating genes, Tables 6a and 6b show that of cell growth-relating genes, Table 7 shows that of cell proliferation-relating genes, Tables 8a and 8b show that of immue response-relating genes, and Table 9 shows that of metastasis-relating genes.













TABLE 3









Exp. pattern
Total















Veh. vs
Veh. vs
relative
t-test/


Gene
Genbank ID
Nm23
CP-Nm23
ratio
p-value















Caspase 14
NM_009809
1.21
4.19
3.46
0.82/0.01 


CD5 antigen
NM_007650
1.21
3.06
2.53
0.20/0.006


PERP, TP53 apoptosis
NM_022032
1.04
2.51
2.41
0.72/0.01 


effector


CD28 antigen
NM_007642
0.81
2.70
3.32
0.29/7.79E−14


ELL associated factor 2
NM_134111
0.72
3.85
5.33
0.08/0.009


Interferon gamma
NM_008337
0.76
5.73
7.55
0.33/0.006


Lectin, galactose
NM_008496
0.79
8.83
11.16
0.33/0.006


binding, soluble 7


Programmed cell death 1
NM_008798
0.98
4.74
4.85
0.85/0.008


Transformation related
NM_011641
2.61
8.22
3.15
0.04/0.009


protein 63


Deoxyribonuclease 1-like 3
NM_007870
1.83
7.37
4.03
0.023/0.006 


Fas apoptotic inhibitory
NM_026976
0.47
1.40
2.96
0.02/0.08 


molecule 3


Interleukin 7
NM_008371
0.82
2.36
2.86
0.028/0.24 


Granzyme B
NM_013542
0.28
0.72
2.52
0.014/0.09 


Cell death-inducing
NM_178373
0.64
2.70
4.21
0.01/0.05 


DFFA-like effector c


Cell death-inducing DNA
NM_007702
0.88
2.43
2.77
0.016/0.31 


fragmentation factor,


alpha subunit-like


effector A




















TABLE 4









Exp. pattern
Total















Veh. vs
Veh. vs
relative
t-test/


Gene
Genbank ID
Nm23
CP-Nm23
ratio
p-value















Glycosylation dependent cell
NM_008134
0.92
4.48
4.88
0.82/0.04


adhesion molecule 1


Selectin, platelet
NM_011347
0.94
3.79
4.04
0.83/0.01


CD226 antigen
NM_178687
0.89
2.57
2.89
0.50/0.01


Mucin 4
NM_080457
0.93
2.05
2.19
0.58/0.02


Cell adhesion molecule with
NM_007697
1.26
5.70
4.54
 0.21/0.002


homology to L1CAM


RGM domain family, member B
BC030405
0.67
3.41
5.11
0.33/0.01


Leupaxin
NM_134152
0.76
2.11
2.80
 0.1/0.02


Integrin, alpha E, epithelial-
NM_008399
0.82
2.66
3.24
0.39/0.02


associated


Coagulation factor VIII
NM_007977
0.85
2.08
2.43
0.47/0.03


Corneodesmosin
NM_001008424
1.87
7.66
4.09
 0.21/0.006


Plakophilin 1
NM_019645
8.26
28.39
3.44
 0.02/0.005


Stabilin 2
NM_138673
0.40
0.80
1.98
0.038/0.221


Myeloid/lymphoid or mixed
AK016557
0.30
0.64
2.12
0.017/0.071


lineage-leukemia translocation


to 4 homolog


Kit ligand
NM_013598
0.20
0.47
2.38
0.008/0.013


Glycoprotein (transmembrane)
NM_053110
0.39
1.03
2.64
0.018/0.814


nmb


Hairy and enhancer of split 1
NM_008235
0.44
0.98
2.23
0.022/0.910


CD164 antigen
NM_016898
0.33
0.89
2.74
0.012/0.377


Procollagen, type XVII, alpha 1
NM_007732
0.36
1.19
3.26
0.017/0.243


Cysteine rich protein 61
NM_010516
0.42
1.14
2.68
0.017/0.327


Cartilage oligomeric matrix
NM_016685
3.53
0.66
0.19
0.012/0.127


protein


Cadherin-like 26
NM_198656
2.85
0.85
0.30
0.017/0.298


Aggrecan
NM_007424
2.84
0.97
0.34
0.013/0.787


Poliovirus receptor-related 4
NM_027893
2.03
0.97
0.48
0.023/0.758


calsyntenin 1
NM_023051
2.17
1.03
0.47
0.020/0.802


laminin, beta 2
NM_008483
2.10
1.05
0.50
0.022/0.665


dermatopontin
NM_019759
3.11
1.22
0.39
0.012/0.187


EGF-like repeats and discoidin
NM_010103
0.83
0.37
0.45
0.522/0.039


I-like domains 3




















TABLE 5









Exp. pattern
Total















vs
vs
relative
t-test/


Gene
Genbank ID
Nm23
CP-Nm23
ratio
p-value





Cell division cycle
NM_026560
0.42
2.63
6.23
0.265/0.043


associated 8


Cyclin B1
NM_172301
0.50
2.23
4.46
0.143/0.031


Dedicator of cytokinesis 4
NM_172803
0.66
2.71
4.13
0.174/0.018


Budding uninhibited by
NM_009773
0.73
2.95
4.07
 0.2/0.016


benzimidazoles 1 homolog,


beta


Dual specificity phosphatase 1
NM_013642
0.76
2.58
3.39
0.113/0.015


Cell division cycle
NM_013538
0.68
2.23
3.27
0.071/0.019


associated 3


FBJ osteosarcoma oncogene
NM_010234
0.98
3.44
3.50
0/0


Protamine 1
NM_013637
0.96
2.19
2.28
0.915/0.027


FBJ osteosarcoma oncogene B
NM_008036
0.98
3.44
3.50
0.881/0.011


Interleukin 1 beta
NM_008361
0.91
2.22
2.45
0.063/0   


M-phase phosphoprotein 1
XM_193936
0.40
0.84
2.09
 0.03/0.258


Bridging integrator 1
NM_009668
0.29
1.02
3.47
0.012/0.866


Jun-B oncogene
NM_008416
0.43
1.00
2.34
0.017/0.977


Calmodulin 1
NM_009790
0.30
1.22
4.11
 0.01/0.189


Stratifin
NM_018754
2.68
1.03
0.38
0.014/0.883


Avian erythroblastosis virus
NM_133659
2.10
0.49
0.23
0.022/0.023


E-26 (v-ets) oncogene related




















TABLE 6a









Exp. pattern
Total















Veh. vs
Veh. vs
relative
t-test/


Gene
Genbank ID
Nm23
CP-Nm23
ratio
p-value















Keratin 5
NM_027011
2.71
8.04
2.96
0.015/0.006


Myosin, heavy polypeptide 8,
AK081482
1.45
2.91
2.00
 0.2/0.02


skeletal muscle, perinatal


Paired box gene 9
NM_011041
1.58
4.49
2.84
0.059/0.009


Troponin T3, skeletal, fast
NM_011620
2.74
6.65
2.43
0.007/0.007


Serine (or cysteine) peptidase
AK003650
1.04
9.02
8.71
 0.26/0.006


inhibitor, clade B, member 3C


B-cell leukemia/lymphoma 2
NM_007535
1.08
2.43
2.26
0.538/0.011


related protein A1c


OVO homolog-like 1
NM_019935
1.08
2.26
2.09
0.553/0.014


Junctophilin 2
NM_021566
1.54
14.08
9.12
0.115/0.006


Myosin, light polypeptide 1
NM_021285
1.30
2.65
2.05
0.127/0.014


Histocompatibility 2, class II
NM_207105
1.23
3.27
2.66
0.101/0.011


antigen A, beta 1


Troponin I, skeletal, slow 1
NM_021467
0.96
2.24
2.33
0.814/0.025


Growth arrest specific 7
NM_008088
0.94
3.02
3.22
0.619/0.012


Tumor necrosis factor (ligand)
NM_009403
0.91
2.89
3.16
0.719/0.016


superfamily, member 8


Frizzled homolog 10
NM_175284
0.99
2.31
2.33
0.946/0.020


Chemokine (C—X—C motif)
NM_018866
1.02
5.12
5.02
0.854/0.008


ligand 13


RAB27A, member RAS oncogene
NM_023635
0.71
2.24
3.15
0.326/0.033


family


Tumor necrosis factor,
NM_025566
0.74
2.15
2.88
0.122/0.021


alpha-induced protein 8-like 1


RAB32, member RAS oncogene
NM_026405
0.71
2.41
3.38
 0.08/0.017


family


Tumor necrosis factor,
NM_009396
0.74
2.37
3.22
0.096/0.017


alpha-induced protein 2





















TABLE 6b







Tumor necrosis factor receptor
NM_011608
0.65
4.42
6.81
0.083/0.009


superfamily, member 17


V-maf musculoaponeurotic
NM_010658
0.83
2.03
2.45
0.203/0.023


fibrosarcoma oncogene family,


protein B (avian)


Interleukin 7 receptor
NM_008372
0.82
2.36
2.86
0.195/0.017


Fgfr1 oncogene partner
NM_201230
0.49
1.15
2.33
0.031/0.328


Tescalcin
NM_021344
0.50
1.16
2.34
0.025/0.272


Burkitt lymphoma receptor 1
NM_007551
0.42
1.93
4.60
0.031/0.031


Protein phosphatase 1,
AK082372
0.18
0.40
2.22
0.019/0.184


regulatory (inhibitor) subunit 14c


Eyes absent 4 homolog
NM_010167
1.07
0.46
0.43
0.721/0.046


Sema domain, transmembrane
AK052232
1.12
0.48
0.43
0.367/0.022


domain (TM), and cytoplasmic


domain, (semaphorin) 6D


Mutated in colorectal cancers
AK086823
0.87
0.41
0.47
0.347/0.019


Sema domain, immunoglobulin
AK049580
0.95
0.38
0.40
0.713/0.18


domain (Ig), short basic domain,


secreted, (semaphorin) 3E


Sine oculis-related homeobox 2
NM_011380
4.18
1.60
0.38
0.022/0.094


homolog


Early B-cell factor 3
AK220542
4.49
1.70
0.38
0.008/0.049


V-abl Abelson murine leukemia
NM_009595
3.90
1.35
0.35
0.021/0.201


viral oncogene 2 (arg,


Abelson-related gene)


Early B-cell factor 2
NM_010095
2.88
1.30
0.45
0.015/0.152


HtrA serine peptidase 3
NM_030127
3.50
1.41
0.40
0.011/0.089


Troponin T2, cardiac
NM_011619
2.33
1.13
0.49
0.018/0.34 


Developmental pluripotency
NM_025274
2.30
0.85
0.37
0.027/0.414


associated 5


Receptor tyrosine kinase-like
NM_013845
2.33
1.06
0.46
0.022/0.654


orphan receptor 1


Phospholipase C, gamma 1
AF027185
3.05
0.99
0.33
0.012/0.951


Deleted in lung and esophageal
AK045848
2.34
0.95
0.41
0.018/0.641


cancer 1


Palate, lung, and nasal
NM_011126
3.68
0.14
0.04
0.007/0/007


epithelium carcinoma associated




















TABLE 7









Exp. pattern
Total















Veh. vs
Veh. vs
relative
t-test/


Gene
Genbank ID
Nm23
CP-Nm23
ratio
p-value





Protein kinase, cAMP
NM_008923
0.87
2.02
2.31
0.494/0.049


dependent regulatory, type


I beta


B-cell translocation gene 1,
NM_007569
0.90
2.02
2.25
0.406/0.024


anti-proliferative


Fc receptor, IgG, low
NM_010187
0.77
2.16
2.82
0.112/0.018


affinity IIb


Immunoglobulin heavy chain
AJ294737
0.71
2.11
2.97
0.079/0.021


6 (heavy chain of IgM)


Neutrophil cytosolic factor 1
NM_010876
1.02
2.35
2.30
0.888/0.019


complement factor B
NM_008198
1.12
3.27
2.91
0.365/0.011


B-cell translocation gene 2,
NM_007570
0.30
0.68
2.28
0.548/0.02 


anti-proliferative


Signal transducer and
NM_009284
0.48
2.34
4.88
 0.04/0.019


activator of transcription 6


Protein tyrosine
NM_011210
0.48
1.35
2.79
0.022/0.1 


phosphatase, receptor type, C


Protein-kinase,
NM_028410
0.31
0.83
2.71
0.011/0.266


interferon-inducible double


stranded RNA dependent


inhibitor, repressor of


(P58 repressor)


Pleckstrin homology domain
AK045134
0.70
0.25
0.36
 0.08/0.011


containing, family K member 1




















TABLE 8a









Exp. pattern
















Veh.
Veh.
Total





Vs
Vs CP-
Relative
t-test/


Gene
Genbank ID
Nm23
Nm23
ratio
p-value















Immunoglobulin
AF296432
0.86
16.25
18.85
0.905/


heavy chain




0.011


(J558 family)







Immunoglobulin
BC004786
0.96
15.09
15.69
0.736/


heavy chain complex




0.006


Histocompatibility 2, T
NM_010397
1.00
2.87
2.89
0.966/


region locus 22




0.013


Lymphocyte
NM_010745
0.99
2.42
2.44
0.936/


antigen 86




0.016


Defensin
NM_054074
0.97
2.19
2.26
0.863/


beta 6




0.025


Immunoglobulin
NM_152839
0.84
26.49
31.61
0.219/


joining chain




0.005


Histocompatibility
NM_010382
0.93
3.65
3.92
0.55/


2, class II




0.01


antigen E beta







Lymphocyte antigen
NM_008530
1.04
3.53
3.41
0.784/


6 complex, locus F




0.01


Thymus cell antigen
NM_009382
1.04
2.47
2.38
0.762/


1, theta




0.016


CD8 antigen,
BC030679
0.78
6.62
8.51
0.142/


alpha chain




0.007


Chemokine
NM_009917
0.86
2.65
3.09
0.265/


(C - C motif)




0.014


receptor 5







Proteoglycan 2,
NM_008920
0.88
3.64
4.13
0.373/


bone marrow




0.01


Histocompatibility
NM_010388
0.93
2.68
2.90
0.518/


2, class




0.014


II, locus Mb2







Defensin beta 3
NM_013756
1.21
5.89
4.89
0.364/







0.007


Histocompatibility
NM_010388
1.08
2.79
2.57
0.53/


2, class II,




0.013


locus Mb2







Histocompatibility
NM_010389
1.06
2.45
2.31
0.647/


2, O region




0.017


beta locus







Histocompatibility
NM_010381
1.11
4.12
3.73
0.423/


2, class




0.009


II antigen E alpha







Transporter 1,
NM_013683
1.21
3.38
2.80
0.226/


ATP - binding




0.011


cassette, sub - family B







(MDR/TAP)







Peptidoglycan
NM_009402
1.25
2.71
2.17
0.159/


recognition protein 1




0.014


T - cell receptor beta,
BC030075
0.60
2.08
3.47
0.148/


variable 13




0.012


Indoleamine - pyrrole
NM_008324
0.68
5.91
8.68
0.079/


2,3 dioxygenase




0.007


Histocompatibility
NM_010387
0.83
2.20
2.64
0.214/


2, class II, locus Mb1




0.019





















TABLE 8b







CD3 antigen, gamma
NM_009850
0.68
5.05
7.46
0.063/0.008


polypeptide


CD96 antigen
NM_032465
0.79
2.39
3.02
0.148/0.017


Histocompatibility 2, class
NM_010378
0.72
2.75
3.82
 0.12/0.014


II antigen A, alpha


Histocompatibility 2, 0
NM_008206
0.65
2.81
4.31
0.053/0.013


region alpha locus


Chemokine (C-X-C motif)
NM_009910
0.63
3.25
5.12
0.048/0.008


receptor 3


CD8 antigen, beta chain 1
NM_009858
0.75
2.15
2.86
0.107/0.021


CD86 antigen
NM_019388
0.83
2.29
2.77
0.066/0.01 


CD3 antigen, delta polypeptide
NM_013487
0.56
3.42
6.16
0.031/0.011


Interferon gamma inducible protein 47
NM_008330
0.67
2.23
3.33
 0.06/0.019


Immunoglobulin lambda chain,
AK008094
0.40
2.85
7.15
0.044/0.015


variable 1


CD180 antigen
NM_008533
0.42
2.19
5.22
0.107/0.042


Coiled-coil domain
AK087049
1.00
0.44
0.44
0.984/0.019


containing 85A


Cathelicidin antimicrobial
NM_009921
1.19
0.37
0.31
0.243/0.016


peptide


Neutrophilic granule protein
NM_008694
1.41
0.36
0.25
0.083/0.007




















TABLE 9









Exp. pattern
Total















Veh. vs
Veh. vs
relative
t-test/


Gene
Genbank ID
Nm23
CP-Nm23
ratio
p-value





Fascin homolog 1, actin
NM_007984
1.31
3.56
2.72
0.118/0.01 


bundling protein


Prostaglandin-endoperoxide
NM_011198
0.81
2.09
2.59
0.459/0.047


synthase 2


Vascular cell adhesion
NM_011693
0.79
1.62
2.05
0.146/0.044


molecule 1









As described in Table 3 above, in case of the apoptosis-relating genes, while the expressions of Caspase 14, cell death-inducing DFFA-like effector c (Cidec), cell death-inducing DNA fragmentation factor and alpha subunit-like effector A (Cidea) were up-regulated by about 3.5-, 4.0-, 2.5- and 2.5-fold, respectively, in the mouse group treated with the cell permeable Nm23 recombinant protein compared to that treated with the control protein.


As described in Table 4 above, in case of the cell adhesion-relating genes, the expression of cadherin-like 26 was down-regulated by about 3.0-fold in the mouse group treated with the cell permeable Nm23 recombinant protein compared to that treated with the control protein.


As described in Table 5 above, in case of the cell cycle regulation-relating genes, the expression of Avian erythroblastosis virus E-26 (v-ets) oncogene was down-regulated by about 4.0-fold in the mouse group treated with the cell permeable Nm23 recombinant protein compared to that treated with the control protein.


As described in Tables 6a and 6b above, in case of the cell growth-relating genes, while the expression of member 17 of a tumor necrosis factor receptor superfamily was up-regulated by about 6.8-fold, the expressions of palate, lung and nasal epithelium carcinoma associated genes were down-regulated by about 26.0-fold in the mouse group treated with the cell permeable Nm23 recombinant protein compared to that treated with the control protein.


As described in Table 7 above, in case of the cell proliferation-relating genes, the expression of signal transducer and activator of transcription 6 was up-regulated by about 5-fold in the mouse group treated with the cell permeable Nm23 recombinant protein compared to that treated with the control protein.


As described in Tables 8a and 8b above, in case of immune response-relating genes, the expressions of immunoglobulin heavy chain (J558 family), immunoglobulin heavy chain complex and immunoglobulin joining chain were up-regulated by about 18-, 15- and 30-fold, respectively, in the mouse group treated with the cell permeable Nm23 recombinant protein compared to that treated with the control protein.


As described in Table 9 above, in case of metastasis-relating genes, the expressions of fascin homolog 1 (actin bundling protein), prostaglandin-endoperoxide synthase 2 and vascular cell adhesion molecule 1 were up-regulated by about 2.5-, 2.5- and 2.0-fold, respectively, in the mouse group treated with the cell permeable Nm23 recombinant protein compared to that treated with the control protein.


Although the invention has been described in detail for the purpose of illustration, it is understood that such detail is solely for that purpose, and variations can be made therein by those skilled in the art without departing from the spirit and scope of the invention which is defined by the following claims.


INDUSTRIAL APPLICABILITY

The cell permeable Nm23 recombinant proteins of the present invention can induce the KSR phosphorylation and inactivation and inhibit the Ras-mediated MAPK cascade by efficiently introducing a metastasis suppressor Nm23 into a cell. Therefore, the cell permeable Nm23 recombinant proteins of the present invention can be effectively used as an anti-metastatic agent capable of preventing cancer metastasis by suppressing the proliferation, differentiation and migration of cancer cells.

Claims
  • 1. A cell permeable Nm23 recombinant protein comprising: a macromolecule transduction domain(MTD) fused to the N-terminus or C-terminus of a human metastasis suppressor Nm23, wherein the MTD comprises SEQ ID NO: 6 or 8, and the Nm23 comprises SEQ ID NO: 2.
  • 2. A cell permeable Nm23 recombinant protein comprising: a first MTD fused to the N-terminus of a human metastasis suppressor Nm23 and a second MTD fused to the C-terminus of said human metastasis suppressor, wherein the first and second MTDs comprise SEQ ID NO: 6 or 8 and the Nm23 comprises SEQ ID NO: 2.
  • 3. The cell permeable Nm23 recombinant protein according to claim 1, wherein the recombinant protein is selected from the group consisting of: a recombinant protein wherein the MTD comprises SEQ ID NO:6 and is fused to the N-terminus of the Nm23; a recombinant protein wherein the MTD comprises SEQ ID NO:6 and is fused to the C-terminus of the Nm23; a recombinant protein wherein the MTD comprises SEQ ID NO: 8 and is fused to the N-terminus of the Nm23; and a recombinant protein wherein the MTD comprises SEQ ID NO:8 and is fused to the C-terminus of the Nm23.
  • 4. The cell permeable Nm23 recombinant protein according to claim 1, wherein the recombinant protein has an amino acid sequence selected from the group consisting of SEQ ID NOS: 28, 30, 32, and 34.
  • 5. A polynucleotide encoding the cell permeable Nm23 recombinant protein according to claim 1.
  • 6. The polynucleotide according to claim 5, wherein the polynucleotide has a nucleotide sequence selected form the group consisting of SEQ ID NOS: 27, 29, 31, and 33.
  • 7. An expression vector comprising the polynucleotide according to claim 5.
  • 8. The expression vector according to claim 7, wherein the expression vector is selected from the group consisting of pET28a(+)-HM2N, pET28a(+)-HNM2, and pET28(+)-HM3N, pET28a(+) -HNM.
  • 9. A transformant comprising the expression vector according to claim 7.
  • 10. The transformant according to claim 9, wherein the transformant is E. coli DH5α/HM3Nm23 (Accession No. KCTC- 11380BP).
  • 11. The transformant according to claim 9, wherein the transformant is E. coli DH5α/HNm23M3 (Accession No. KCTC- 11381 BP).
  • 12. A method of producing a cell permeable Nm23 recombinant protein comprising culturing the transformant according to claim 1 or 2.
  • 13. The cell permeable Nm23 recombinant protein according to claim 1 or 2, further comprising: a nuclear localization sequence(NLS) and a histidine-tag affinity domain, said nuclear localization sequence and histidine-tag affinity domain being covalently coupled to one end of the recombinant protein.
  • 14. The cell permeable Nm23 recombinant protein of claim 2 wherein the first and second MTDs comprise SEQ ID NO 8.
  • 15. The cell permeable Nm23 recombinant protein according to claim 2, wherein the recombinant protein has an amino acid sequence of SEQ ID NO: 36.
  • 16. A polynucleotide encoding the cell permeable Nm23 recombinant protein according to claim 2.
  • 17. The polynucleotide according to claim 16, wherein the polynucleotide has a nucleotide sequence of SEQ ID NO: 35.
  • 18. An expression vector comprising the polynucleotide according to claim 16.
  • 19. The expression vector according to claim 18, wherein the expression vector is pET28(+)-HM3NM3.
  • 20. A transformant comprising the expression vector according to claim 18.
  • 21. A pharmaceutical composition comprising the cell permeable Nm23 recombinant protein according to claim 1 or 2 as an effective ingredient and a pharmaceutically acceptable carrier, which if effective for preventing metastasis by inhibiting proliferation, differentiation or migration of cancer cells.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a National Stage of PCT/KR08/005221 filed Sep. 4, 2008 and claims the benefit of U.S. 60/969,714 filed Sep. 4, 2007.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/KR2008/005221 9/4/2008 WO 00 3/3/2010
Publishing Document Publishing Date Country Kind
WO2009/031835 3/12/2009 WO A
Foreign Referenced Citations (9)
Number Date Country
2 336 223 Aug 2001 CA
1033405 Sep 2000 EP
1 127 576 Aug 2001 EP
WO 9105793 May 1991 WO
WO 9534295 Dec 1995 WO
WO 9811232 Mar 1998 WO
WO 0175067 Oct 2001 WO
WO 02083179 Oct 2002 WO
WO 2008093982 Aug 2008 WO
Non-Patent Literature Citations (22)
Entry
Hiroshi Harada, et al., “Antitumor Protein Therapy; Application of the Protein Transduction Domain to the Development of a Protein Drug for Cancer Treatment”, Breast Cancer, Japanese Breast Cancer Society, vol. 13, No. 1, XP-002544010, Jan. 1, 2006, pp. 16-26.
Alvaro Leone, et al., “Reduced Tumor Incidence, Metastatic Potential, and Cytokine Responsiveness of nm23-Transfected Melanoma Cells”, Cell, Cell Press, vol. 65, No. 1, XP002326572, Apr. 5, 1991, pp. 25-35.
Extended European Search Report issued on Feb. 4, 2011 in the corresponding European Application No. 08829132.3.
Anna D'Angelo, et al., “Prune cAMP Phosphodiesterase Binds nm23-H1 and Promotes Cancer Metastasis”, Cancer Cell, vol. 5, No. 2, Feb. 2004, pp. 137-149.
Daewoong Jo, et al., “Epigenetic Regulation of Gene Structure and Function with a Cell-Permeable Cre Recombinase”, Nature Biotechnology, vol. 19, Oct. 2001, pp. 929-933.
Daewoong Jo, et al., “Intracellular Protein Therapy with SOCS3 Inhibits Inflammation and Apoptosis”, Nature Medicine, vol. 11, No. 8, Aug. 2005, pp. 892-898.
Qing Lin, et al., “Enhanced Cell-Permeant Cre Protein for Site-Specific Recombination in Cultured Cells”, BMC Biotechnology, Oct. 22, 2004, pp. 1-13.
Mike S. S. Chang, et al., “Dissecting Intracellular Signaling Pathways with Membrane-Permeable Peptides”, Science STKE, vol. 47, pl1. [DOI: 10.1126/stke.2000.47.pl1], Aug. 29, 2000, pp. 1-10.
Nicholas J. MacDonald, et al., “A Serine Phosphorylation of Nm23, and Not Its Nucleoside Diphosphate Kinase Activity, Correlates with Suppression of Tumor Metastatic Potential”, The Journal of Biological Chemistry, vol. 268, No. 34, Issue of Dec. 5, Received for Publication on May 20, 1993, pp. 25780-25789.
Patricia S. Steeg, “Breast Cancer Advocacy and Basic Research: A Scientist's Perspective”, Breast Disease, 10(5,6), 1998, pp. 47-50.
Fabio Gervasi, et al., “nm23 Influences Proliferation and Differentiation of PC12 Cells in Response to Nerve Growth Factor”, Cell Growth and Differentiation, vol. 7, Dec. 1996, pp. 1689-1695.
Renzo Hirayama, et al., “Positive Relationship Between Expression of Anti-metastatic Factor (nm23 Gene Product or Nucleoside Diphosphate Kinase) and Good Prognosis in Human Breast Cancer”, Journal of the National Cancer Institute, Brief Communications, vol. 83, No. 17, Sep. 4, 1991, pp. 1249-1253.
Toshihiro Nakayama, et al., “Expression in Human Hepatocellular Carcinoma of Nucleoside Diphosphate Kinase, a Homologue of the nm23 Gene Product”, Journal of the National Cancer Institute, Reports , vol. 84. No. 17, Sep. 2, 1992, pp. 1349-1354.
Alvaro Leone, et al., “Transfection of Human nm23-H1 into the Human MDA-MB-435 Breast Carcinoma Cell Line: Effects on Tumor Metastatic Potential, Colonization and Enzymatic Activity”, Oncogene, 8, 1993, pp. 2325-2333.
Alvaro Leone, et al., “Reduced Tumor Incidence, Metastatic Potential, and Cytokine Responsiveness of nm23-Transfected Melanoma Cells”, Cell, vol. 65, Apr. 5, 1991, pp. 25-35.
Alvaro Leone, et al., “Evidence for nm23 RNA Overexpression, DNA Amplification and Mutation in Aggressive Childhood Neuroblastomas”, Oncogene, 8, 1993, pp. 855-865.
Nicholas J. MacDonald, et al., “Site-directed Mutagenesis of nm23-H1”, vol. 271, No. 41, Issue of Oct. 11, The Journal of Biological Chemistry, Received for Publication on Mar. 11, 1996, pp. 25107-25116.
Deborah K. Morrison, et al., “KSR: a MAPK Scaffold of the Ras Pathway?”, Journal of Cell Science, 114 (9), 2001, pp. 1609-1612.
W Richard Burack, et al., “Signal Transduction: Hanging on a Scaffold”, Current Opinion in Cell Biology, 12, 2000, pp. 211-216.
Tony Pawson, et al., “Signaling Through Scaffold, Anchoring, and Adaptor Proteins”, American Association for the Advancement of Science, Science, New Series, vol. 278, No. 5346, Dec. 19, 1997, pp. 2075-2080.
Toru Yoshida, et al., “Microsatellite Instability in Gallbladder Carcinoma: Two Independent Genetic Pathways of Gallbladder Carcinogenesis”, Journal of Gastroenterology, 35, 2000, pp. 768-774.
Melanie T. Hartsough, et al., “Nm23-H1 Metastasis Suppressor Phosphorylation of Kinase Suppressor of Ras via a Histidine Protein Kinase Pathway”, The Journal of Biological Chemistry, vol. 277, No. 35, Issue of Aug. 30, Received for Publication on Apr. 1, 2002, pp. 32389-32399.
Related Publications (1)
Number Date Country
20100323971 A1 Dec 2010 US
Provisional Applications (1)
Number Date Country
60969714 Sep 2007 US