Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL MARKERS FOR DOPAMINERGIC NEURON PROGENITOR CELLS
Document Type and Number:
WIPO Patent Application WO/2013/015457
Kind Code:
A1
Abstract:
The present invention provides a method for selecting dopaminergic neuron progenitor cells, which comprises detecting any one or more of markers selected from the group consisting of CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA-4, CD49f, SERINC4, CCR9, PHEX, TMPRSS11E, HTR1E, SLC25A2, Ctxn3, Ccl7, Chrnb4, Chrna3, Kcnv2, Grm2, Syt2, Lim2, Mboat1, St3gal6, Slc39a12, Tacr1, Lrtm1, Dscam and CD201.

Inventors:
TAKAHASHI JUN (JP)
DOI DAISUKE (JP)
SAMATA BUMPEI (JP)
ONO YUICHI (JP)
Application Number:
PCT/JP2012/069785
Publication Date:
January 31, 2013
Filing Date:
July 27, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV KYOTO (JP)
EISAI R&D MAN CO LTD (JP)
TAKAHASHI JUN (JP)
DOI DAISUKE (JP)
SAMATA BUMPEI (JP)
ONO YUICHI (JP)
International Classes:
C12N5/0797
Domestic Patent References:
WO2007119759A12007-10-25
WO2011007900A12011-01-20
Other References:
EDMAN,L.C. ET AL.: "The beta-chemokines CCL2 and CCL7 are two novel differentiation factors for midbrain dopaminergic precursors and neurons.", EXP. CELL RES., vol. 314, no. 10, 10 June 2008 (2008-06-10), pages 2123 - 2130, XP022714950
DEZAWAM ET AL., J CLIN INVEST, vol. 113, 2004, pages 1701 - 1710
VIERBUCHEN T ET AL., NATURE, vol. 463, 2010, pages 1035 - 1041
O. FUMITAKA ET AL., NAT. BIOTECHNOL, vol. 26, 2008, pages 215 - 224
E. KROON ET AL., NAT BIOTECHNOL., vol. 26, 2008, pages 443 - 452
M. KANATSU-SHINOHARA ET AL., BIOL. REPROD., vol. 69, 2003, pages 612 - 616
K. SHINOHARA ET AL., CELL, vol. 119, 2004, pages 1001 - 1012
MASANORI TAKEHASHI ET AL.: "Experimental Medicine", vol. 26, 2008, YODOSHA, pages: 41 - 46
T. WAKAYAMA ET AL., SCIENCE, vol. 292, 2001, pages 740 - 743
S. WAKAYAMA ET AL., BIOL. REPROD., vol. 72, 2005, pages 932 - 936
J. BYRNE ET AL., NATURE, vol. 450, 2007, pages 497 - 502
J.B. CIBELLI ET AL., NATURE BIOTECHNOL., vol. 16, 1998, pages 642 - 646
SAYAKA WAKAYAMA ET AL.: "Experimental Medicine", vol. 26, 2008, pages: 47 - 52
SUEMORI H ET AL., BIOCHEM
KAWASAKI H ET AL., NEURON, vol. 28, 2000, pages 31 - 40
ONO Y ET AL., DEVELOPMENT, vol. 134, 2007, pages 3213 - 3225
EIRAKU M ET AL., CELL STEM CELL, vol. 3, 2008, pages 519 - 532
See also references of EP 2737057A4
Attorney, Agent or Firm:
KAWAGUCHI, Yoshiyuki et al. (4-10 Higashi Nihonbashi 3-chome, Chuo-k, Tokyo 04, JP)
Download PDF:
Claims:
CLAIMS

1. A method for producing dopaminergic neuron progenitor cells, said method comprising: extracting dopaminergic neuron progenitor cells from a cell population comprising dopaminergic neuron progenitor cells by using as an index/indices the positivity/positivities of any one or more of markers selected from the group consisting of CD 15 (SSEA-1 ), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184,

DisalogangUosid GD2, SSEA-4, CD49f, SERINC4, CCR9, PHEX, TMPRSS1 IE, HTR1E, SLC25A2, Ctxn3, Ccl7, Chmb4, Chma3, Kcnv2, Gim2, Syt2, Lim2, Mboatl, St3gal6, Slc39al 2, Tacr 1 , Lrtml and Dscam, and/or the negativity of CD201.

2. The method according to claim 1 , wherein the positive marker is Lrtml .

3. The method according to claim 1 or 2, wherein said step further comprises extracting dopaminergic neuron progenitor cells by using as an index/indices the positivity/positivities of corin and/or Lmx la

4. The method according to any one of claims 1 to 3, wherein said dopaminergic neuron progenitor cells are human dopaminergic neuron progenitor cells.

5. The method according to any one of claims 1 to 4, wherein said cell population comprising dopaminergic neuron progenitor cells is a cell population whose differentiation is induced from pluripotent stem cells, or a cell population composed of cells of an isolated tissue(s). 6. The method according to claim 5, wherein said cell population whose differentiation is induced from pluripotent stem cells is a cell population obtained by culturing pluripotent stem cells in a medium supplemented with a BMP inhibitor and a TGFp inhibitor.

7. A method for detecting dopaminergic neuron progenitor cells in a cell population comprising dopaminergic neuron progenitor cells, said method comprising detecting

dopaminergic neuron progenitor cells using as an index/indices the positivity/positivities of any one or more of markers selected from the group consisting of CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184, DisalogangUosid GD2, SSEA-4, CD49f, SERINC4, CCR9, PHEX, TMPRSS1 IE, HTR1E, SLC25A2, Ctxn3, Ccl7, ChrnM, Chrna3, Kcnv2, Grm2, Syt2, Lim2, Mboatl, St3gal6, Slc39al2, Tacrl, Lrtml and Dscam, and/or the negativity of CD201.

8. The method according to claim 7, wherein the positive marker is Lrtml .

9. The method according to claim 7 or 8, wherein said step further comprise detecting dopaminergic neuron progenitor cells by using as an index/indices the positivity positivities of corin and/or Lmxl

10. The method according to any one of claims 7 to 9, wherein said dopaminergic neuron progenitor cells are human dopaminergic neuron progenitor cells.

11. The method according to any one of claims 7 to 10, wherein said cell population comprising dopaminergic neuron progenitor cells is a cell population whose differentiation is induced from pluripotent stem cells, or a cell population composed of cells of an isolated tissue(s).

12. The method according to claim 11 , wherein said cell population whose differentiation is induced from pluripotent stem cells is a cell population obtained by culturing pluripotent stem cells in a medium supplemented with a BMP inhibitor and a TGFP inhibitor.

13. A kit for detecting dor)arninergic neuron progenitor cells, comprising a reagent(s) for detecting any one or more of markers selected from the group consisting of CD 15 (SSEA- 1 ), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA , CD49f, SERTNC4, CCR9, PHEX, TMPRSS1 IE, HTRIE, SLC25A2, Ctxn3, Ccl7, Chrnb4, Chma3, Kcnv2, Grm2, Syt2, Lim2, Mboatl, St3gal6,

Slc39al2, Tacrl, Lrtml, Dscam and CD201.

14. The kit according to claim 13, wherein said reagent(s) is are an antibody/antibodies.

15. The kit according to claim 14, wherein the antibody is anti-Lrtml antibody which recognized an extracellular domain of Lrtml.

16. A therapeutic agent for Parkinson's disease, comprising dopaminergic neuron progenitor cells produced by the method according to any one of claims 1 to 6.

Description:
DESCRIPTION

NOVEL MARKERS FOR DOPAMINERGIC NEURON PROGENITOR CELLS BACKGROUND ART

[0001]

Parkinson's disease is a neurodegenerative disease caused by loss of dopaminergic neural cells in the mesencephalic substantia nigra, and about 4 million people in the world are currently suffering from this disease. For treatment of Parkinson' s disease, pharmacotherapy with L-dopa or a dopamine agonist; the coagulation method or deep brain stimulation by stereotaxy; fetal mesencephalic grafting; or the like has been carried out

[0002]

Fetal mesencephalic grafting is problematic from an ethical point of view because of its source of supply, and the risk of infection is high in this treatment Thus, a therapeutic method using neural cells induced from pluripotent stem cells such as embryonic stem cells (ES cells) or induced pluripotent stem cells (iPS cells) has been proposed (Wernig M, et al., Proc Natl Acad Sci U S A.2008, 105: 5856-5861). However, the possibility of formation of a benign tumor after transplantation of induced neural cells has been pointed out thus, selection of safe cells that can survive has been demanded for the transplantatioa

[0003]

In view of this, genes that can be used as markers for dopaminergic neural cells and/or dopaminergic neuron progenitor cells have been reported (WO2005/052190, WO 2006/009241 and WO 2007/119759), but it is considered that more markers are necessary for restrictive extraction of specific cells suitable for transplantation.

SUMMARY OF THE INVENTION

[0004]

The present invention aims to extract dopaminergic neuron progenitor cells from a cell population comprising dopaminergic neuron progenitor cells. Therefore, an object of the present invention is to provide markers specific for dopaminergic neuron progenitor cells.

[0005] In order to solve the above-described object, the inventors of the present invention focused attention on corin, which is a cell surface membrane protein that has been considered to be a marker of dopaminergic neuron progenitor cells, and extracted corin-positive cells from a cell population comprising neural progenitor cells or neural cells produced by differentiation induction from pluripotent stem cells. The inventors then discovered genes and glycolipids as cell surface markers specifically expressed in these cells, and confirmed that dopaminergic neuron progenitor cells can be obtained by using these markers as indices, thereby completed the present invention.

[0006]

An aspect of the present invention is to provide a method for producing dopaminergic neuron progenitor cells, said method comprising:

extracting dopaminergic neuron progenitor cells from a cell population comprising dopaminergic neuron progenitor cells by using as an index/indices the positivity/posiuvines of any one or more of markers selected from the group consisting of CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184,

Disalogangliosid GD2, SSEA-4, CD49f, SERINC4, CCR9, PHEX, TMPRSS1 IE, HTR1E, SLC25A2, Ctxn3, Ccl7, Chrnb4, Chrna3, Kcnv2, Grm2, Syt2, Lim2, Mboatl, St3gal6, Slc39al2, Tacrl , Lrtml and Dscam, and/or the negativity of CD201.

Another aspect of the present invention is to provide the method as described above , wherein the positive marker is Lrtml.

Another aspect of the present invention is to provide the method as described above, wherein said step of extracting dopaminergic neuron progenitor cells further uses as an index/indices the positivity/positivities of corin and/or Lmxla.

Another aspect of the present invention is to provide the method as described above, wherein said dopaminergic neuron progenitor cells are human dopaminergic neuron progenitor cells.

Another aspect of the present invention is to provide the method as described above, wherein said cell population comprising dopaminergic neuron progenitor cells is a cell population whose differentiation was induced from pluripotent stem cells, or a cell population composed of cells of an isolated tissue(s).

Another aspect of the present invention is to provide the method as described above, wherein said cell population whose differentiation was induced from pluripotent stem cells is a cell population obtained by culturing pluripotent stem cells in a medium supplemented with a BMP inhibitor and a TGFp inhibitor.

Another aspect of the present invention is to provide a method for detecting

dopaminergic neuron progenitor cells in a cell population comprising dopaminergic neuron progenitor cells, said method comprising detecting dopaminergic neuron progenitor cells using as an index/indices the positivity/positivities of any one or more of markers selected from the group consisting of CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA- , CD49f, SERINC4, CCR9, PHEX, TMPRSSl IE, HTRIE, SLC25A2, Ctxn3, Ccl7, ChmM, Chma3, Kcnv2, Grm2, Syt2, Lim2, Mboatl, St3gal6, Slc39al2, Tacrl, Lrtml and Dscam, and/or the negativity of CD201.

Another aspect of the present invention is to provide the method as described above, wherein the positive marker is Lrtml .

Another aspect of the present invention is to provide the method as described above, wherein said step of detecting dopaminergic neuron progenitor cells further uses as an index/indices the positivity/positivities of corin and/or Lmxla

Another aspect of the present invention is to provide the method as described above, wherein said dopaminergic neuron progenitor cells are human dopaminergic neuron progenitor cells.

Another aspect of the present invention is to provide the method as described above, wherein said cell population comprising dopaminergic neuron progenitor cells is a cell population whose differentiation was induced from pluripotent stem cells, or a cell population composed of cells of an isolated tissue(s).

Another aspect of the present invention is to provide the method as described above, wherein said cell population whose differentiation was induced from pluripotent stem cells is a cell population obtained by culturing pluripotent stem cells in a medium supplemented with a BMP inhibitor and a TGFp inhibitor.

Another aspect of the present invention is to provide a kit for detecting dopaminergic neuron progenitor cells, comprising a reagents) for detecting any one or more of markers selected from the group consisting of CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57,

CD58, CD59, CD81, CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA-4, CD49f, SERLNC4, CCR9, PHEX, TMPRSS1 IE, HTRIE, SLC25A2, Ctxn3, Ccl7, Chrn , Chrna3, Kcnv2, Grm2, S t2, Lim2, Mboatl, St3gal6, Slc39al2, Tacrl, Lrtml, Dscam and CD201.

Another aspect of the present invention is to provide the kit as described above, wherein said reagent(s) is/are an antibody/antibodies.

Another aspect of the present invention is to provide the kit as described above, wherein the antibody is anti-Lrtml antibody which recognized an extracellular domain of Lrtml .

Another aspect of the present invention is to provide a therapeutic agent for Parkinson's disease, comprising dopaminergic neuron progenitor cells produced by the method as described above.

BRIEF DESCRIPTION OF THE DRAWINGS

οη

[Fig. 1] Fig. 1 shows fluorescent image for immunostaining (photograph). Fig. 1A shows the image of mouse Lrtml positive and negative cells just after sorting. Lmxla, DAPI and Corin are shown in red, blue and green, respectively. Fig. IB shows the image of mouse Lrtml positive and negative cells at 7 days after sorting. In the left panel TH and DAPI are shown in green and blue, in the second panel fiom left TH and Tuj 1 are shown in green and white, in the third panel from left Nurrl and TH are shown in red and green, and in right panel DAT and DAPI are shown in green and blue, respectively.

[Fig.2] Fig.2 A shows PCR analysis for mouse Lrtml positive and negative cells just after sorting (photograph). Fig.2B shows growth chart of the cells after sorting.

[Fig. 3] Fig. 3 shows fluorescent image for irnmunostaining (photograph). Fig. 3A shows the image of human LRTMl positive and negative cells just after sorting. LMXla, DAPI and CORIN are shown in red, blue and green, respectively. Fig. 3B shows the image of human LRTMl positive and negative cells at 7 days after sorting. LMXla, DAPI, NURRl and TH are shown in red, blue, white and green, respectively.

DESCRIPTION OF THE EMBODIMENTS OF THE INVENTION

[0008]

The present invention will now be described below detail.

[0009]

The present invention relates to: a method for producing dopaminergic neuron progenitor cells, the method comprising: extracting dopaminergic neuron progenitor cells from a cell population comprising dopaminergic neuron progenitor cells by using as an index/indices the posiuvity/positivities of any one or more of markers selected from the group consisting of CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA-4, CD49f, SERJNC4, CCR9, PHEX, TMPRSS1 IE, HTR1E, SLC25A2, Ctxn3, Ccl7, Chmb4, Chrna3, Kcnv2, Grm2, Syt2, Lim2, Mboatl, St3gal6, Slc39al2, Tacrl, Lrtml and Dscam, and/or the negativity of CD201 ; and

a method for detecting dopaminergic neuron progenitor cells in a cell population comprising dopaminergic neuron progenitor cells, the method comprising detecting

dopaminergic neuron progenitor cells using as an index/indices the positivity/positivities of any one or more of markers selected from the group consisting of CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA-4, CD49f, SERINC4, CCR9, PHEX, TMPRSS1 IE, HTRIE, SLC25A2, Ctxn3, Ccl7, Chmb4, Chma3, Kcnv2, Grm2, Syt2, Lim2, Mboatl, St3gal6, Slc39al2, Tacrl, Lrtml and Dscam, and/or the negativity of CD201.

[0010]

In the present invention, the term "dopaminergic neuron progenitor cells" means cells which are characterized in that they are positive for at least one of corin (WO2006/009241 ) and Lmxl a (W 02005/052190)) and which become dopaminergic neural cells after maturatioa The dopaminergic neural cells are not restricted, and may be characterized in that they are positive for tyrosine hydroxylase (TH). However, in the present invention, dopaminergic neuron progenitor cells and dopaminergic neural cells are not clearly distinguished from each other, and TH-positive cells can also be said to be dopaminergic neuron progenitor cells.

[0011]

In the present invention, the origin of the cell population comprising dopaminergic neuron progenitor cells is not restricted as long as the cell population is a population of cells comprising dopaminergic neuron progenitor cells. For example, the origin may be a cell population contained in a tissue obtained by an arbitrary method, or a cell line established from a tissue. The tissue herein is a brain tissue, preferably fetal midbrain. The cell population may also be a cell population comprising neural progenitor cells obtained by differentiation induction ofbone marrow stromal cells (DezawaM, etal., J Clin Invest 2004, 113: 1701-1710) or pluripotent stem cells, or a cell population comprising neural progenitor cells directly induced from fibroblasts (Vierbuchen T, et al., Nature.2010, 463: 1035-1041).

[0012]

In the present invention, the term "extraction of dopaminergic neuron progenitor cells" means to increase the ratio of dopaminergic neuron progenitor cells relative to the ratios of other types of cells, and preferably means to concentrate dopaminergic neuron progenitor cells to a content of not less than 50%, not less than 60%, not less than 70%, not less than 80% or not less than 90%. The above term more preferably means to obtain a cell population which comprises dopaminergic neuron progenitor cells at a content of 100%.

[0013]

The markers to be used in the present invention, CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA-4, CD49f, SERINC4, CCR9, PHEX, TMPRSS1 IE, HTRIE, SLC25A2, Ctxn3, Ccl7, ChrnM, Chma3, Kcnv2, Grm2, Syt2, Lim2, Mboatl, St3gal6, Slc39al2, Tacrl, Lrtml, Dscam and CD201 , include the polynucleotides represented by the NCBI accession numbers described in Table 1 and proteins encoded thereby, and transcriptional variants, splicing variants, homologues and fragments thereof. The fragments herein preferably correspond to extracellular domains of the markers, in consideration of the fact that the markers are used for recognition of cells. In this invention, preferable marker is Lrtm 1. More preferably, t he marker is the f agment of extracellular domain of Lrtml . The marker can be used together with conventional marker(s), such as corin(WO2006/009241), Lmxla(WO2005/052190), 65B13 (WO2004/038018) and 185A5 (WO2007/119759).

[0014]

Pable 1]

NCBI Accession number

Marker

Mouse Homo sapiens

CD15 (SSEA-1) NM 010242 NM 002033

CD24 NM 009846 NM 013230

NM 002389 NM 153826 NM 172350 NM 172351

CD46 NM_010778

NM 172352 NM 172353 NM 172359 NM_172361

NM 001777

CD47 NM_010581

NMJ98793

CD49b NM 008396 NM 002203

NM 018644

CD57 NM_029792

NM 054025

NM 001779

CD58 - NM_001144822

NM 203330 NM 000611 NM 203329

NM 001111060 NM 203331

CD59

NM_007652 NM 001127223

NM 001127225 NM 001127226 NM_001127227

CD81 NM 133655 NM 004356

CD90 NM 009382 NM 006288

NM 001012662

NM 001161413 NM 002394

CD98

NM 008577 NM 001012664

(SLG3A2, SLC7A5)

NM 011404 NM 001013251

NM 003486

NM 001728

NM 001077184

CD147 NM 198589

NM 009768

NMJ98591

NM 001008540

CD184 NM_009911

NM_003467

NM 000210

CD49f NM_008397

NM_001079818

CD201 NM 011171 NM 006404

SERINC4 NM 001025371 NM 001033517

NM 001166625 NM 006641

CCR9

NM 009913 NM_031200

PHEX NM 011077 NM 000444 TMPRSS11E NM 172880 NM 014058

HTR1E - NM 000865

SLC25A2 NM 001159275 NM 031947

NM 001048252

Ctxn3 NM_001134697

NM 001127385

Ccl7 NM 013654 NM 006273

Chmb4 NM 148944 NM 000750

NM 000743

Chma3 NMJ45129

NM_001166694

cnv2 NMJ83179 NMJ33497

NM 000839

Grm2 NM_001160353

NM_001130063

NM 001136504

Syt2 NM_009307

NM_177402

NM 001161748

Lim2 NMJ77693

NM_030657

Mboatl NM_153546 NM 00I080480

St3gal6 NM_018784 NM_006100

Slc39al2 NM_001012305 NM_001145195

NM 001058

Tacrl NM_009313

NM 015727

Lrtml NM_176920 NM_020678

Dscam NM_031174 NM_001389

[0015]

Disalogangliosid GD2 and SSEA-4, which are used as markers, are glycolipids. Disalogangliosid GD2 is a sphingoglycolipid which is represented as Cer-Glc-Gal(NeuAc- NeuAc)-GalNAc, wherein Cer represents ceramide, Glc represents glucopyranose, Gal represents galactopyranose, NeuAc represents acetylneuramic acid, and GalNAc represents acetylgalactopyranose. SSEA is a sphingoglycolipid having sialosyl-galactosyl-globoside (sialosyl-Gb5) as an epitope.

[0016]

<Pluripotent Stem Cells>

In cases where a cell population comprising dopaminergic neuron progenitor cells is prepared by differentiation of pluripotent stem cells, the pluripotent stem cells have pluripotency which enables the cells to differentiate into any cells existing in the living body, and also have growth ability. Examples of the pluripotent stem cells include, but are not limited to, embryonic stem (ES) cells, embryonic stem cells derived from a cloned embryo obtained by nuclear transfer ("ntES cells"), germline stem cells ("GS cells"), embryonic germ cells ("EG cells"), induced pluripotent stem (iPS) cells, and pluripotent cells derived from cultured fibroblasts and bone marrow stem cells (Muse cells). The pluripotent stem cells are preferably ES cells, ntES cells or iPS cells.

[0017]

(A) Embryonic Stem Cells

ES cells are stem cells established from the inner cell mass of an early embryo (for example, blastocyst) of a mammal such as human or mouse, which cells have pluripotency and growth ability by self-renewal.

[0018]

ES cells are stem cells originated from the inner cell mass of a blastocyst which is the embryo formed following die 8-cell stage and the morula stage of a fertilized egg, and ES cells have ability to differentiate into any cells constituting an adult, that is, the so called pluripotency of differentiation, and growth ability by self-renewal. ES cells were discovered in mouse in 1981 (M. J. Evans and M. H. Kaufinan (1981), Nature 292:154-156), and followed by establishment of ES cell lines of primates such as human and monkey (J. A. Thomson et al. (1998), Science 282:1145-1147; J. A. Thomson et al. (1995), Proc. Natl. Acad. Sci. USA, 92:7844-7848; J. A. Thomson et al. (1996), Biol. Reprod., 55:254-259; J. A. Thomson and V. S. Marshall (1998), Curr. Top. Dev. Biol., 38: 133-165).

[0019]

ES cells can be established by removing the inner cell mass from the blastocyst of a fertilized egg of a subject animal, followed by culturing the inner cell mass on fibroblasts as feeders. The cells can be maintained by subculturing using a medium supplemented with a substance(s) such as leukemia inhibitory factor (LIF) and/or basic fibroblast growth factor (bFGF). Methods of establishment and maintenance of human and monkey ES cells are described in, for example, US 5,843,780 B; Thomson JA, et al. (1995), Proc Natl. Acad. Sci. U S A.92:7844-7848; Thomson JA, et al. (1998), Science.282:1145-1147; H. Suemori et al. (2006), Biochem. Biophys. Res. Commua, 345:926-932; M. Ueno et al. (2006), Proc. Natl. Acad. Sci. USA, 103:9554-9559; H. Suemori et al. (2001), Dev. Dyn., 222:273-279; H. Kawasaki et al. (2002), Proc. Natl. Acad. Sci. USA, 99:1580-1585; and Klimanskaya I, et al. (2006), Nature. 444:481-485.

[0020]

In terms of the medium for preparation of ES cells, human ES cells can be maintained, for example, using D E /F- 12 medium supplemented with 0.1 mM 2-mercaptoethanol, 0.1 mM non-essential amino acids, 2 mM L-glutamic acid, 20% KSR and 4 ng/ml bFGF at 37°C under a moist atmosphere of 2% C0 2 /98% air (O. Fumitaka et al. (2008), Nat Biotechnol., 26:215-224). Further, ES cells need to be subcultured every 3 to 4 days, and the subculture can be carried out using 0.25% trypsin and 0.1 mg/ml collagenase IV in PBS supplemented with 1 mM CaCl 2 and20% KSR

[0021]

Selection of ES cells can be generally carried out by the Real-Time PCR method using as an index indices expression of a gene marker(s) such as alkaline phosphatase, Oct-3/4 and/or Nanog. In particular, for selection of human ES cells, expression of a gene marker(s) such as OCT-3/4, NANOG and/or ECAD can be used as an index/indices (E. Kroon et al. (2008), Nat Biotechnol, 26:443-452).

[0022]

In terms of human ES cell lines, for example, WA01(H1) and WA09(H9) can be obtained from WiCell Research Institute, and KhES-1 , KhES-2 and KhES-3 can be obtained from Institute for Frontier Medical Sciences, Kyoto University (Kyoto, Japan).

[0023]

(B) Germline Stem Cells

Germline stem cells are pluripotent stem cells derived from testis, and play a role as the origin for spermatogenesis. Similarly to ES cells, these cells can be induced to differentiate into various series of cells, and, for example, have a property to enable preparation of a chimeric mouse by transplantation of the cells to a mouse blastocyst (M. Kanatsu-Shinohara et al. (2003)

Biol. Reprod., 69:612-616; K. Shinoharaet al. (2004), Cell, 119:1001-1012). Germline stem cells are capable of self-renewal in a medium containing glial cell line-derived neurotrophic factor (GDNF), and, by repeating subculture under the same culture conditions as those for ES cells, germline stem cells can be obtained (Masanori Takehashi et al. (2008), Experimental Medicine, 26(5) (extra edition):41 -46, Yodosha (Tokyo, Japan)).

[0024] (C) Embryonic Germ Cells

Embryonic germ cells are established from fetal primordial germ cells and have pluripotency similarly to ES cells. They can be established by culturing primordial germ cells in the presence of substances such as LIF, bFGF and stem cell factor (Y. Matsui et al. (1992), Cell, 70:841-847; J. L. Resnicketal. (1992), Nature, 359:550-551).

[0025]

(D) Induced Pluripotent Stem Cells

Induced pluripotent stem (iPS) cells can be prepared by introducing specific reprogramming factors to somatic cells, which reprogiamming factors are in the form of DNA or protein. iPS cells are somatic cell-derived artificial stem cells having properties almost equivalent to those of ES cells, such as pluripotency of differentiation and growth ability by self- renewal (K. Takahashi and S. Yamanaka (2006) Cell, 126:663-676; K. Takahashi et al. (2007), Cell, 131:861-872; J. Yuetal. (2007), Science, 318:1917-1920;Nakagawa, M. etal.,Nat.

Biotechnol.26:101-106 (2008); WO 2007/069666). The reprogramming factors may be constituted by genes or gene products thereof, or non-coding RNAs, which are expressed specifically in ES cells; or genes or gene products thereof, non-coding RNAs or low molecular weight compounds, which play important roles in maintenance of the undifferentiated state of ES cells. Examples of the reprogramming factors include Oct3/4, Sox2, Soxl , Sox3, Soxl 5, Soxl7, Klf4, Klf2, c-Myc,N-Myc, L-Myc, Nanog, Lin28, Fbxl5, ERas, ECAT15-2, Tell, beta- catenin, Lin28b, Salll, Sall4, Esrrb, Nr5a2, Tbx3 and Glisl, and these reprogramming factors may be used either alone or in combination. Examples of the combinations of the

reprogramming factors include those described in WO2007/069666; WO2008/118820;

WO2009/007852; WO2009/032194; WO2009/058413; WO2009/057831; WO2009/075119; WO2009/079007; WO2009/091659; WO2009/101084; WO2009/101407; WO2009/102983; WO2009/114949; WO2009/117439; WO2009/126250; WO2009/126251; WO2009/126655; WO2009/157593; WO2010/009015; WO2010/033906; WO2010/033920; WO2010/042800; WO2010/050626; WO 2010/056831; WO2010/068955; WO2010/098419; WO2010/102267; WO 2010/111409; WO 2010/111422; WO2010/115050; WO2010/124290; WO2010/147395; WO2010/147612; Huangfu D, et al. (2008), Nat. Biotechnol., 26: 795-797; Shi Y, et al. (2008), Cell Stem Cell, 2: 525-528; Eminli S, et al. (2008), Stem Cells.26:2467-2474; Huangfu D, et al.

(2008), Nat Biotechnol.26:1269-1275; Shi Y, et al. (2008), Cell Stem Cell, 3, 568-574; Zhao Y, et al. (2008), Cell Stem Cell, 3:475-479; Marson A, (2008), Cell Stem Cell, 3, 132-135; Feng B, et al. (2009), Nat Cell Biol. 11 : 197-203; R.L. Judson et al., (2009), Nat. Biotech., 27:459-461 ; Lyssiotis CA, et al. (2009), ProcNatl Acad Sci U S A. 106:8912-8917; Kim JB, et al. (2009), Nature.461 :649-643; Ichida JK, et al. (2009), Cell Stem Cell. 5:491-503; Heng JC, et al. (2010), Cell Stem Cell.6:167-74; Han J, et al. (2010), Nature.463:1096-100; Mali P, et al. (2010), Stem Cells.28:713-720; and Maekawa M, et al. (2011), Nature.474:225-9.

[0026]

Examples of the above-described reprograrnming factors also include histone deacetylase (HDAC) inhibitors [for example, low molecular weight inhibitors such as valproic acid (VPA), trichostatin A, sodium butyrate, MC 1293 and M344; and nucleic acid-type expression inhibitors such as siRNAs and shRNAs against HDAC (e.g., HDAC1 siRNA Smartpool® (Millipore) and HuSH 29mer shRNA Constructs against HDAC1 (OriGene))], MEK inhibitors (for example, PD184352, PD98059, U0126, SL327 and PD0325901), Glycogen synthase kinase-3 inhibitors (for example, Bio and CHIR99021), DNA

methyltransferase inhibitors (for example, 5'-azacytidine), histone methyltransferase inhibitors (for example, low molecular weight inhibitors such as ΒΓΧ-01294; and nucleic acid-type expression inhibitors such as siRNAs and shRNAs against Suv39hl, Suv39h2, SetDBl and G9a), L-channel calcium agonists (for example, Bayk8644), butyric acid, TGF inhibitors or ALK5 inhibitors (for example, LY364947, SB431542, 616453 and A-83-01), p53 inhibitors (for example, siRNAs and shRNAs against p53), ARID3 A inhibitors (for example, siRNAs and shRNAs against ARTD3A), miRNAs such as miR-291-3p, miR-294, miR-295 and mir-302, Wnt Signaling (for example, soluble Wnt3a), neuropeptide Y, prostaglandins (for example, prostaglandin E2 and prostaglandin J2), hTERT, SV40LT, UTFl , IRX6, GLIS1, PITX2 and DMRTBl, which are employed for enhancing the establishment efficiency, and, in the present description, these factors employed for the purpose of enhancement of the establishment efficiency are not particularly distinguished from the above-described reprograrnming factors.

[002η .

In cases where the reprograrnming factors are in the form of protein, the reprograrnming factors may be introduced into somatic cells by a method such as lipofection, fusion with a cell membrane-permeable peptide (e.g., HTV-derived TAT or polyarginine), or microinjection.

[0028] In cases where the reprogramming factors are in the form of DNA, the r^rogramrning factors may be introduced into somatic cells by a method such as use of a vector including virus, plasmid and artificial chromosome vectors; lipofection; use of liposome; or microinjectioa Examples of the virus vectors include retrovirus vectors, lentivirus vectors (these are described in Cell, 126, pp. 663-676, 2006; Cell, 131, pp. 861-872, 2007; and Science, 318, pp. 1917-1920, 2007), adenovirus vectors (Science, 322, 945-949, 2008), adeno-associated virus vectors and Sendai virus vectors (WO 2010/008054). Examples of the artificial chromosome vectors include human artificial chromosomes (HACs), yeast artificial chromosomes (YACs), and bacterial artificial chromosomes (BACs and PACs). Examples of the plasmids include plasmids for mammalian cells (Science, 322:949-953, 2008). The vectors may contain a regulatory sequence(s) such as a promoter, enhancer, ribosome binding sequence, terminator and/or polyadenylation site; and, as required, a sequence of a selection marker such as a drug resistance gene (e.g., kanamycin-resistant gene, ampicillin-resistant gene or puromycin-resistant gene), thymidine kinase gene or diphtheria toxin gene; a gene sequence of a reporter such as the green-fluorescent protein (GFP), β-glucuronidase (GUS) or FLAG; and/or the like to enable expression of the nuclear reprogramming factors. Further, in order to remove, after introduction of the above vector into somatic cells, the genes encoding the reprogramming factors, or both the promoters and the genes encoding the reprogramming factors linked thereto, the vector may have LoxP sequences upstream and downstream of these sequences.

[0029]

Further, in cases where the reprogramming factors are in the form of RNA, each r^rogramming factor may be introduced into somatic cells by a method such as lipofection or microinjection, and an RNA into which 5-methylcytidine and pseudouridine (TriLink

Biotechnologies) were incorporated may be used in order to suppress degradation (Warren L, (2010) Cell Stem Cell.7:618-630).

[0030]

Examples of the medium for induction of the iPS cells include the DMEM, DMEM/F 12 and DME media supplemented with 10 to 15% FBS (these media may further contain LIF, pemcillin/streptomycin, puromycin, L-glutamine, non-essential amino acids, β-mercaptoethanol and/or the like, as appropriate); and commercially available media [for example, a medium for culturing mouse ES cells (TX-WES medium, Thromb-X), medium for culturing primate ES cells (medium for primate ES/iPS cells, ReproCELL) and serum-free medium (mTeSR,

Stemcell Technology)].

[0031]

Examples of the culture method include a method wherein somatic cells and

reprogramming factors are brought into contact with each other at 37°C in the presence of 5%

CO2 in DMEM or DMEM F12 medium supplemented with 10% FBS, and the cells are cultured for about 4 to 7 days, followed by plating the cells on feeder cells (e.g., mitomycin C-treated STO cells or SNL cells) and starting culture in a bFGF-containing medium for culturing primate ES cells about 10 days after the contact between the somatic cells and the reprogramming factors, thereby allowing iPS-like colonies to appear about 30 to about 45 days after the contact, or later.

[0032]

Alternatively, the cells may be cultured at 37°C in the presence of 5% CO2 on feeder cells (e.g., mitomycin C-treated STO cells or SNL cells) in the DMEM medium supplemented with 10% FBS (this medium may further contain LIF, penicillin/streptomycin, puromycin, L- glutamine, non-essential amino acids, β-mercaptoethanol and/or the like, as appropriate) for about 25 to about 30 days or longer, to allow ES-like colonies to appear. Preferred examples of the culture method include a method wherein the somatic cells themselves to be reprogrammed are used instead of the feeder cells (Takahashi K, et al. (2009), PLoS One.4:e8067 or

WO2010/137746), and a method wherein an extracellular matrix (e.g., Laminin-5

(WO2009/123349) or Matrigel (BD)) is used instead.

[0033]

Other examples of the culture method include a method wherein culture is carried out using a serum-ftee medium (Sun N, et al. (2009), Proc Natl Acad Sci U S A. 106: 15720-15725). Further, in order to enhance the establishment efficiency, iPS cells may be established under low oxygen conditions (at an oxygen concentration of not less than 0.1 % and not more than 15%)

(Yoshida Y, et al. (2009), Ceil Stem Cell. 5:237-241 or WO2010/013845).

[0034]

During the culture, the medium is replaced with a fresh medium once every day from Day 2 of the culture. The number of the somatic cells used for nuclear reprogramming is not restricted, and usually within the range of about 5 χ 10 3 to about 5 χ 10 6 cells per 100-cm 2 area on the culture dish. [0035]

iPS cells may be selected based on the shape of each formed colony. In cases where a drug resistance gene was introduced as a marker gene to be expressed in conjunction with a gene that is expressed when a somatic cell was reprogrammed (e.g., Oct3/4 or Nanog), established iPS cells can be selected by culturing the cells in a medium containing the corresponding drug (selection medium). Further, iPS cells can be selected by observation under a fluorescence microscope in cases where the marker gene is the gene of a fluorescent protein; by adding a luminescent substrate in cases where the marker gene is the gene of luciferase; or by adding a coloring substrate in cases where the marker gene is the gene of a coloring enzyme.

[0036]

The term "somatic cells" used in the present description means any animal cells

(preferably cells of mammals including human) excluding germ-line cells and totipotent cells such as eggs, oocytes and ES cells. Examples of the somatic cells include, but are not limited to, any of fetal somatic cells, neonatal somatic cells, and mature, healthy and diseased somatic cells, as well as any of primary cultured cells, subcultured cells and established cell lines. Specific examples of the somatic cells include (1) tissue stem cells (somatic stem cells) such as neural stem cells, hematopoietic stem cells, mesenchymal stem cells and dental pulp stem cells; (2) tissue progenitor cells; and (3) differentiated cells such as lymphocytes, epithelial cells, endothelial cells, muscle cells, fibroblasts (skin cells and the like), hair cells, hepatic cells, gastric mucosal cells, enterocytes, spleen cells, pancreatic cells (pancreatic exocrine cells and the like), brain cells, lung cells, kidney cells and adipocytes.

[0037]

In cases where iPS cells are used as a material for cells to be transplanted, somatic cells whose HLA genotype is the same or substantially the same as that of the individual to which the cells are to be transplanted are preferably used in view of prevention of the rejection reactioa

The term "substantially the same" herein means that the HLA genotype is matching to an extent at which the immune reaction against the transplanted cells can be suppressed with an immunosuppressive agent For example, the somatic cells have matched HLA types at the 3 loci HLA-A, HLA-B and HLA-DR, or at the 4 loci further including HLA-C.

[0038]

(E) ES Cells Derived from Cloned Embryo Obtained by Nuclear Transfer ntES cells are ES cells derived from a cloned embryo prepared by the nuclear transfer technique, and have almost the same properties as those of ES cells derived from fertilized eggs (T. Wakayama et al. (2001), Science, 292:740-743; S. Wakayama et al. (2005), Biol. Reprod, 72:932-936; J. Byrne et al. (2007), Nature, 450:497-502). That is, an ntES (nuclear transfer ES) cell is an ES cell established from the inner cell mass of a blastocyst derived from a cloned embryo obtained by replacement of the nucleus of an unfertilized egg with the nucleus of a somatic cell. For preparation of an ntES cell, the combination of the nuclear transfer technique (J.B. Cibelli et al. (1998), Nature Biotechnol., 16:642-646) and the ES cell preparation technique (described above) is employed (Sayaka Wakayama et al. (2008), Experimental Medicine 26(5) (extra edition), pp.47-52). In nuclear tramfer, reprogramming can be achieved by injecting the nucleus of a somatic cell into a mammalian enucleated unfertilized egg and culturing the resultant for several hours.

[0039]

(F) Mullilineage-differentiating Stress Enduring Cells (Muse Cells)

Muse cells are pluiipotent stem cells produced by the method described in

WO2011/007900. More specifically, Muse cells are cells having pluripotency obtained by subjecting fibroblasts or bone marrow stromal cells to trypsin treatment for a long period, preferably to trypsin treatment for 8 hours or 16 hours, followed by suspension culture of the treated cells. Muse cells are positive for SSEA-3 and CD105.

[0040]

<Method for Preparing Cell Population Comprising Dopaminergic neuron Progenitor Cells from Pluripotent Stem Cells>

The method of differentiation induction of pluripotent cells into a cell population comprising dopaminergic neuron progenitor cells is not restricted, and examples of the method include the followings.

[0041]

Pluripotent stem cells are separated by an arbitrary method and subjected to suspension culture, or adherent culture using a coated culture dish. In the method of separation, the cells may be mechanically separated, or may be separated using an EDTA solution (e.g. 0.5 niM EDTA solution or Versene (Invitrogen)), separation solution having protease activity and collagenase activity (e.g., CTK (collagenase-trypsin-KSR) solution (Suemori H, et al., Biochem , Biophys Res Commua 345: 926-32, 2006), Accutase (TM) or Accumax (TM)), or separation solution having only collagenase activity. In the suspension culture, a culture dish having a surface which has not been subjected to an artificial treatment for the purpose of enhancing cell adhesiveness such as coating treatment with an extracellular matrix or the like, a culture dish having a surface which has been artificially treated such that adhesion is suppressed (for example, coated with polyhydroxyethylmethacrylate (poly-HEMA)), or a culture dish having a surface treated with Lipidure (NOF Corporation), may be used. In the adherent culture, the culture dish may be one coated with Matrigel (BD), type I collagen, type IV collagen, gelatin, laminin, heparan sulfate proteoglycan or entactin, or a combinations thereof.

[0042]

The adherent culture may be carried out by co-culturing with feeder cells. Examples of the feeder cells used in the co-culture include PA6 cells (Kawasaki H, et al., Neuron.2000, 28: 3140).

[0043]

A medium to be used for culturing animal cells may be prepared as a basal medium.

Examples of the basal medium include GMEM (Glasgow Minimum Essential Medium), IMDM (Iscove's Modified Dulbecco's Medium), Medium 199, Eagle's Minimum Essential Medium (EMEM), aMEM, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium and Fischer's medium, and mixtures of these media. The medium is preferably GMEM. The medium may contain serum, but the medium is preferably serum-free to eliminate heterologous components. In such cases, the medium may contain, as required, one or more serum replacements such as albumin, transferrin, fatty acid, insulin, collagen precursor, trace element, Knockout Serum Replacement (KSR) (serum replacement for FBS in ES cell culture) and/or ITS supplement.

[0044]

The medium may further contain, for promoting survival, growth and differentiation induction of the cells, one or more substances such as 2-mercaptoethanol, thioglycerol, B27 supplement, N2 supplement, lipid, amino acid, L-glutamine, Glutamax (Invitrogen), nonessential amino acid, vitamin, cytokine, hedgehog family, BMP inhibitor, TGFP family inhibitor, Rho kinase inhibitor, Wnt signaling inhibitor, antibiotic, antioxidant, pyruvic acid, retinoic acid, ascorbic acid, buffer and/or inorganic salt, although the substances are not restricted. Examples of the cytokine include neurotrophic factors and fibroblast growth factors (FGFs), and preferred examples of the cytokine include GDNF, BDNF, FGF-2, FGF-8 and FGF-20. Examples of the hedgehog family include sonic hedgehog (SHH). Examples of the BMP inhibitor include protein-based inhibitors such as Chordin, Noggjn and Follistatin; Dorsomorphin (6-[4-(2- piperidin-l-yl-ethoxy)phenyl]-3-pyridin-4-yl-pyiazolo[l ,5-a]pyrimidine) and its derivatives (P. B. Yu et al. (2007), Circulation, 116:Π_60; P.B. Yu et al. (2008), Nat. Chem. Biol, 4:33^1; J. Hao et al. (2008), PLoS ONE, 3(8):e2904); and LDN-193189 (4-(6-(4-(piperazin-l- yl)phenyl)pyrazDlo[l,5-a]pyrimidin-3-yl)quinoline). Dorsomorphinand LDN-193189 are commercially available, and can be obtained from Sigma-Aldrich and Stemgent, respectively. Examples of the TGFP family inhibitor include Lefly- 1 (e.g, NCBI Accession Nos.

NM 010094 (mouse) andNM_020997 (human)); SB431542 and SB202190 (these are described in R. K. Lindemann et al., Mol. Cancer, 2003, 2:20); SB505124 (GlaxoSmithKline); NPC30345, SD093, SD908 and SD208 (Scios); LY2109761, LY364947 and LY580276 (Lilly Research Laboratories); A-83-01 (WO 2009146408); and derivatives thereof. Examples of the Rho kinase inhibitor include Fasudil (l-(5-Isoquinolinesulfonyl)homopiperazine Hydrochloride), Y-27632 ((R)-(+)-trans-N-(4-pyridyl) -(l -anmoelhyl)-cyclohexanecarboxamide · 2HC1 · H 2 0), H-1152 (e.g, Sasaki, et al, Pharmacol. Ther.2002, 93: 225-232) and Wf-536 (e.g, Nakajima, et al. Cancer Chemother Pharmacol.2003, 52(4): 319-324). Examples of the Wnt signaling inhibitor include XAV939 (Shih-Min A. Huang, et al, Nature 461, 614-620, 2009),

Dickkopfl (Dkkl ), insulin-like growth factor-binding protein (IGFBP) (WO2009/131166) and siRNAs against β-catenin.

[0045]

For differentiation induction into a cell population comprising dopaminergic neuron progenitor cells, the medium preferably comprises a BMP inhibitor and TFG β inhibitor.

[0046]

More preferred examples of the medium include GMEM supplemented with a BMP inhibitor, TGFP family inhibitor, Rho kinase inhibitor, KSR, pyruvic acid, non-essential amino acid and 2-mercaptoethanol. In cases where additional agents are added, the agents to be added may be different among the stages of culture. More preferably, the cells are cultured for not less than 4 days in the above-described medium, and then cultured in GMEM supplemented with

KSR, pyruvic acid, non-essential amino acid and 2-mercaptoethanol with further addition, as required, of FGF-2, FGF-8 and FGF-20.

[004η

The culture temperature is not restricted, and preferably about 30 to 40°C, more preferably about 37°C. The culture is carried out under an atmosphere of C0 2 -containing air, wherein the C ( ¾ concentration is preferably about 2 to 5%. The culture is continued for a period necessary for expression of corin and/or Lmxla, and, for example, the culture period is not less than 4 days.

[0048]

The thus produced cell population comprising dopaminergic neuron progenitor cells need not to be a cell population comprising only the single type of cells, and may be a cell population also comprising another/other type(s) of cells.

[0049]

<Method of Extraction or Detection of Dopaminergic neuron Progenitor Cells>

Each of the reagent(s) to be used for extracting or detecting dopaminergic neuron progenitor cells from the cell population comprising dopaminergic neuron progenitor cells is not restricted as long as it has specific affinity to CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA-4, CD49f, SERINC4, CCR9, PHEX, TMPRSS1 IE, HTR1E, SLC25A2, Ctxn3, Ccl7, Chrnb4, Chrna3, Kcnv2, Grm2, Syt2, Lim2, Mboatl, St3gal6, Slc39al2, Tacrl, Lrtml, Dscam, CD201, corin and/or Lmxla. Antibodies, aptamers, peptides, compounds, and the like which specifically recognize the markers may be used. The reagent is preferably an antibody or its fragment.

[0050]

In the present invention, the antibodies may be either a polyclonal or monoclonal antibody. These antibodies can be prepared using a technique well known to those skilled in the art (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley and Sons. Sections 11.12-11.13). More specifically, in cases where each antibody is a polyclonal antibody, the polyclonal antibody can be obtained by immunizing a non-human mammal such as a rabbit with a protein such as CD 15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81 , CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA-4, CD49f, SERINC4, CCR9, PHEX, TMPRSS1 IE, HTR1E, SLC25A2, Ctxn3, Ccl7, ChrnM, ' Chrna3, Kcnv2, Grm2, Syt2, Lim2, Mboatl, St3gal6, Slc39al2, Tacrl, Lrtml, Dscam, CD201, corin or Lmx 1 a, expressed in E. coli, a mammalian cell line or the like and then purified; an oligopeptide having a partial amino acid sequence of such a protein (preferably the fragments corresponding to extracellular domain(s) of the protein); or a glycolipid; and isolating the polyclonal antibody from the serum of the immunized animal according to a conventional method. In cases where an oligopeptide or glycolipid is used for the immunization, it is preferably cross-linked to an appropriate carrier protein to prepare an immunogen. The carrier protein in such cases is not restricted, and keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA) or the like is often used. In this invention, the protein fragment consisting of a part of sequence of SEQ ID NO: 1 or 2 is exemplified as the fragments corresponding to extracellular domain of mouse and human Lrtml .

[0051]

In cases where the antibody is a monoclonal antibody, the monoclonal antibody can be obtained from among hybridoma cells prepared by cell fusion of spleen cells obtained from the above-described immunized non-human mammal with myeloma cells (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley and Sons. Sections 11.4- 11.11). Examples of the fragment of the antibody include a part of the antibody (e.g., Fab fragment) and a synthetic antibody fragment (e.g., single-stranded Fv fragment "ScFv").

Antibody fragments such as the Fab and F(abh fragments can also be prepared by well known methods such as genetic engineering techniques. Examples of the antibody against corin include the antibody described in WO2006/009241. Examples of the antibody against Lmxla include the antibody described in WO2005/052190.

[0052]

Each of the reagents such as antibodies having affinities, which are used for the purpose of recognition or separation of cells expressing CD15 (SSEA-1), CD24, CD46, CD47, CD49b, CD57, CD58, CD59, CD81, CD90, CD98, CD147, CD184, Disalogangliosid GD2, SSEA-4, CD49f, SERLNC4, CCR9, PHEX, TMPRSS1 IE, HTRIE, SLC25A2, Ctxn3, Ccl7, Chmb4, Chma3, Kcnv2, Gim2, Syt2, Lim2, , Mboatl, St3gal6, Slc39al2, Tacrl, Lrtml, Dscam, CD201, corin and/or Lmxla, may be bound or conjugated, for example, to a detectable substance such as a fluorescent label, radioactive label, chemiluminescent label, enzyme, biotin or streptavidin, or to a substance that allows isolation/extraction of the cells, such as protein A, protein G, beads or magnetic beads.

[0053]

The reagent having afiBnity may also be indirectly labeled. The labeling may be carried out by various methods known to those skilled in the art, and examples thereof include a method wherein a preliminarily labeled antibody (secondary antibody) that specifically binds to the antibody is used.

[0054]

Examples of the method for detecting dopaminergic neuron progenitor cells include, but are not limited to, use of a flow cytometer, protein chip or the like.

[0055]

Examples of the method for extracting dorjarninergic neuron progenitor cells include, but are not limited to, a method by conjugating particles to the reagent having affinity, method by sorting cells using magnetic beads by the magnetism (e.g., MACS), method using a fluorescent label to use a cell sorter, and method using a carrier (e.g., cell-concentrating column) to which an antibody or the like is immobilized.

[0056]

<Application to Regenerative Medicine>

The dorjaminergic neuron progenitor cells obtained by the present invention can be effectively used in the field of regenerative medicine for supplying dopaminergic cells which have been lost. Examples of diseases to which the above cells can be applied include

Parkinson's disease.

[0057]

In order to suppress tumorigenesis after transplantation, it is preferred to administer cells that do not express a pluripotency marker(s). Examples of the pluripotency markers) herein include Nanog and Oct3/4.

EXAMPLES

[0058]

The present invention will now be described more specifically by way of Examples below, but, needless to say, the present invention is not restricted to thereto.

[0059] <Example 1>

Human ES cells ( hES-1) were obtained from Institute for Frontier Medical Sciences, Kyoto University (Suemori H, et al. Biochem Biophys Res Commun.345 : 926-32, 2006). Human iPS cells (253G4) were obtained from Center for iPS Cell Research and Application, Kyoto University (Nakagawa M, et al. Nat Biotechnol.26: 101-6, 2008).

[0060]

The human ES cells and human iPS cells were induced to differentiate into neural cells by the SDIA (stromal cell-derived inducing activity) method described in Kawasaki H, et al., Neuron. 2000, 28: 31 -40 which was slightly modified. Briefly, the human ES cells or iPS cells were suspended in CTK (collagenase-trypsin-KSR) solution to form cell clumps (each comprising 10 to 20 cells), which were then plated on PA6 feeders in a 10-cm dish at a ratio of 1 :2. The cells were cultured in GMEM (GIBCO) supplemented with 10 uM Y-27632 (ROCK inhibitor, WAKO), 2 μΜ dorsomorphin (BMP inhibitor, Sigma), 10 μΜ SB431542

(TGFp/Activin/Nodal inhibitor, Sigma), 8% KSR, 1 mM pyruvate (Sigma), 0.1 mM MEM non essential amino acid (NEAA, Invitrogen) and 0.1 mM 2-Mercaptoethanol (2-ME, WAKO) for 4 days. The medium was then replaced with GMEM supplemented with 8% KSR, 1 mM pyruvate, 0.1 mM NEAA and 0.1 mM 2-ME, and the culture was continued for 10 to 17 days.

[0061]

The thus produced cell population comprising dopaminergic neuron progenitor cells were stained using antibodies against human cell surface markers contained in Lyoplate (BD) and an anti-corin antibody (Ono Y, et al., Development 2007, 134: 3213-3225), and analyzed by flow cytometry. As a result, the genes and glycolipids described in Table 2 were identified as cell surface markers expressed on corin-positive cells. Further, the gene described in Table 3 was identified as a cell surface marker which is not expressed on corin-positive cells.

[0062] |Table2]

[0063] [Table 3]

[0064]

<Example 2>

Human ES cells (KhES-1) were obtained from Institute for Frontier Medical Sciences, Kyoto University. Human iPS cells (253G4) were obtained from Center for iPS Cell Research and Application, Kyoto University.

[0065]

The human ES cells and human iPS cells were induced to differentiate into neural cells by the above-described SDIA method and the SFEBq (serum-free embryoid body quick) method described in Eiraku M, et al., Cell Stem Cell.2008, 3: 519-532 which were slightly modified. In the modified SFEBq method, the human ES cells or the human iPS cells were treated with Accumax (Innovate cell technologies) to be suspended into single cells, and then plated on a 96-well plate (Lipidure-coat U96w, Nunc) at 9,000 cells/150 uL/well. The cells were cultured in GMEM supplemented with 10 uM Y-27632, 0.1 uM LDN193186 (BMP inhibitor, STEMGENT), 0.5 uM A-83-01 (TGFp/Activin/Nodal inhibitor, WAKO), 8% KSR, 1 mM pyruvate, 0.1 mM NEAA and 0.1 mM 2-ME for 5 days. The medium was then replaced with GMEM supplemented with 8% KSR, 1 mM pyruvate, 0.1 mM NEAA and 0.1 mM 2-ME, and the culture was continued for 5 to 9 days.

[0066]

From the cell population comprising dopaminergic neuron progenitor cells produced by the modified SDIA method or modified SFEBq method, corin-positive cells and corin-negative cells were sorted with a flow cytometer. By the microarray method, genes specifically showing higher expression in corin-positive cells relative to corin-negative cells were identified. Among the genes which specifically showed high expression, cell surface markers were as shown in Table 4.

[006η [Table 4]

[0068]

<Example 3>

Mouse ES cells in which the GFP gene was knocked-in into the Lmxl a locus and GFP expression is regulated by the endogenous Lmxl a promoter were induced to differentiate into neural cells by the SFEB method. Briefly, the ES cells were suspended in GMEM

supplemented with 2 mM L-Gln, 0.1 mM NEAA, 1 mM Sodium pyruvate, 0.1 mM 2-ME and 5%KSR, and subjected to suspension culture using a 96-well plate at a density of 3,000 cells/150 ul/well. On Day 3 of the suspension culture, 100 ng/ml FGF8 was added to the medium. On the next day, 100 ng/ml SHH was added to the medium. On Day 7 of the suspension culture, the medium was replaced with GMEM supplemented with 100 ng/ml SHH, 20 ng/ml BDNF, 200 nM Ascorbic acid, 2 mM L-Gln, 0.1 mM NEAA, 1 mM Sodium pyruvate and 0.1 mM 2- ME. On Day 11 of the suspension culture, the cells were collected, and 4 fractions, that is, ( 1 ) GFP (Lmxla)-positive and corin-positive; (2) GFP (Lmxla)-positive and corin-negative; (3) GFP (Lmx 1 a)-negative and corin-positive; and (4) GFP (Lmx 1 a)-negative and corin-negative; were separated using a flow cytometer. By the microarray method, genes which were more strongly expressed in the fractions (1 ), (2) and (3) than in the fraction (4) were identified.

Among the thus identified genes specifically expressed in the corin- and/or Lmxl a-positive cells, cell surface markers were as shown in Table 5.

[0069] [Table 5]

[0070]

<Example 4>

Mouse iPS cells (440A-3) were obtained fiom RIKEN BRC (Okita K et al. Science 322: 949-953 (2008)).

[0071]

The mouse iPS cells were induced to differentiate into neural cells by the following SFEBq method;

Day 0: The mouse iPS cells were dissociated into single cells, and then plated on a 96-well plate (Lipidure-coat U96w, Nunc) at 6,000 cells/150 uL/well. The cells were cultured in mSFEB Medium (GMEM supplemented with 2mM L-Glutamine, 1 mM Sodium pyruvate, 0.1 mM NEAA and lmM 2-ME) supplemented with 5% KSR.

Day 1 : Half medium was replaced with mSFEB Medium supplemented with 5% KSR, 200 ng μΐ rhFGF8b (Peprotech) and 200 ng/μΐ rmSHH (R&D).

Day 3: Half medium was replaced with SFEB Medium supplemented with 5% KSR, 100 ng/ul rhFGF8b and 100 ng/μΐ rmSHH.

Day 5: Half medium was replaced with SFEB Medium supplemented with 5% KSR, 100 ng ul rhFGF8b and 100 ng/ul rmSHH.

Day 7: Half medium was replaced with SFEB Medium supplemented with N2, 200 nM

Ascorbic acid and 20 ng/ml rhBDNF.

Day 9: The cells were dissociated into the single cells with Accutase. FACS buffer (PBS supplemented with 20 mM Glucose (Wako) and 2% FBS (Cell culture bioscience)) was added to the cell solution, and then immunostained with monoclonal anti-mLrtml antibodies at 4 degree C for 20min. Stained cells and unstained cells were sorted with FACS AriaH (BD Biosciences), and then plated on 8well chamber (OFL (Poly-L-ornithine, Fibronectin and Laminin)-coat) at 50,000 cells /well. The cells were cultured in mAdhesion Medium

(DMEM F12 supplemented with 2 mM L-Glutamine, N2, B27, 200 nM Ascorbic acid, 20 ng/ml rhBDNF, 10 ng/ml rhGDNF, 1% KSR, Penicillin Streptomycin) supplemented with 30 uM Y-27632. The monoclonal anti-mLrtml antibodies were obtained from the culture supernatant of myeloma fusion cells. The fusion cells were produced with well-known method. Briefly, rat were immunized with isolated protein consisting of the extracellular region encoding sequence in mouse Lrtml gene, and lymphocytes were removed therefrom and fused with myeloma cells. The anti-mLrtm 1 antibody is checked by recognition of the transfectant cells overexpressing mouse Lrtml and mouse embryonic midbrain.

Day 11 : All medium was replaced with mAdhesion Medium.

Day 14: All medium was replaced with mAdhesion Medium.

[0072]

At 0 and 7 days after sorting, the cells were analyzed with PCR and immunostaining.

The result is shown in Fig. 1 and 2. The content rate of each neural marker positive cells is shown in Table 6 and 7. Just after sorting, Lmxl a and Corin positive cells were frequently confirmed in Lrrm 1 positive cells. Similarly, at 7 days after sorting, dopaminergic neuron marker positive cells were f equently confirmed in Lrtml positive cells. Furthermore, the Lrrm 1 positive cells did not proliferate after sorting. These results show that Lrtm 1 can be substituted for Lmxl a and Corin and one of the potent marker for postmitotic dopaminergic neuron progenitor cells.

[0073]

[Table 6] : Day 0 after sorting

[0075]

<Example 5>

Human ES cells (Kh-ESl) were induced to differentiate into neural cells by the following SFEBq method;

Day 0: The human ES cells were treated with Accumax (Innovate cell technologies) to be suspended into single cells, and then plated on a 96-well plate (Lipidure-coat U96w, Nunc) at 6,000 cells/150 uL/well. The cells were cultured in hSFEB Medium (GMEM supplemented with lniM Sodium pyruvate, O.lmMNEAA and ImM 2-ME) supplemented with 8% KSR, 0.1 uM LDN193186, 0.5 μΜ A-83-01 and 30 uM Y-27632,

Day 1 : Half medium was replaced with hSFEB Medium supplemented with 8% KSR, 0.1 μΜ LDN193186, 0.5 uM A-83-01, 30 uM Y-27632, 100 ng/ul rhFGF8b (Peprotech), 100 ng/ul rmSHH (R&D) and 4 uM Purmoroharnine (Wako).

Day 3 : Half medium was replaced with hSFEB Medium supplemented with 8% KSR, 0.1 μΜ LDN193186, 0.5 uM A-83-01, 30 uM Y-27632, 100 ng/ul rhFGF8b, 100 ng/ul rmSHH, 2uM Purmo hamine and 6 μM CHIR99021.

Day 5: Half medium was replaced with hSFEB Medium supplemented with 8% KSR, 0.1 uM LDN193186, 100 ng/μΐ rhFGF8b, 100 ng/ul rmSHH, 2uM Fairmorphamineand 3 uM

CHIR99021.

Day 7: Half medium was replaced with hSFEB Medium supplemented with 8% KSR and 3 uM CHIR99021.

Day 9: Half medium was replaced with hSFEB Medium supplemented with 8% KSR and 3 uM CHIR99021.

Day 12: The cells were dissociated into the single cells with Accutase. FACS buffer (PBS supplemented with 20 mM Glucose (Wako) and 2%0) FBS (Cell culture bioscience)) was added to the cell solution, and then immunostained with monoclonal anti-hLrtml antibodies at 4 degree C for 20min. Stained cells and unstained cells were sorted with F ACSAriall (BD Biosciences), and then plated on 8 well chamber (OL (Poly-L-ormthine and Laminin)-coat) at 50,000 cells /well. The cells were cultured in hAdhesion Medium (Neurobasal supplemented with 2 mM L-Glutamine, B27, 200 nM Ascorbic acid, 20 ng/ml rhBDNF, 10 ng/ml rhGDNF, Penicillin Streptomycin) supplemented with 30 uM Y-27632. The monoclonal anti-hLRTM 1 antibodies were obtained from the culture supernatant of myeloma fusion cells. The fusion cells were produced with well-known method. Briefly, rat were immunized with isolated protein consisting of the extracellular region encoding sequence in human LRTM1 gene, and lymphocytes were removed therefrom and fused with myeloma cells. The anti-hLRTM 1 antibody is checked by recognition of the transfectant cells overexpressing human LRTM1. Day 14: All medium was replaced with hAdhesion Medium.

Day 17: All medium was replaced with hAdhesion Medium.

[0076]

At 0 and 7 days after sorting, the cells were analyzed with iinmunostaining. The result is shown in Fig. 3. The content rate of each neural marker positive cells is shown in Table 8 and 9. At 0 and 7 days after sorting, LMXla positive cells were frequently confirmed in Lrtml positive cells. These results show that LRTMl can be substituted for LMXla. At 7 days after sorting, NURRl positive cells in LRTMl ositive cells were contained in TH positive cells at a relatively high percentage. Lrtml is one of the cell membrane protein. Therefore, alive dopaminergic neuron progenitor cells can be extracted by using the LRTMl as a marker.

[007η

[Table 8] : Day 0 after sorting

Sorted cells by Sorted sells by

LRTMlpositive gate LRTMlnegative gate

Rate of Lmxla positive cells to

52.9% 4.5% DAPI positive nuclei

Rate of Corin positive cells to

54.3% 40.5% DAPI positive nuclei

Rate of Corin and Lmxla

positive cells to DAPI positive 37.5% 0.2% nuclei

[0078]

[Table 9] : Day 7 after sorting

Sorted cells by Sorted sells by

LRTMlpositive gate LRTMlnegative gate

Rate of Lmxla positive cells to

22.3% 11.0% DAPI positive nuclei

Rate of Nurrl positive cells to

20.5% 14.6% DAPI positive nuclei

Rate of Tyrosine Hydroxylase

(TH) positive cells to DAPI 12.6% 12.9% positive nuclei

Rate of Lmxla positive cells to

63.2% 24.4% TH positive cells

Rate of Nurrl positive cells to

65.8% 28.9% TH positive cells