Conditionally Immortalized Long-term Stem Cells And Methods Of Making And Using Such Cells

CAMBIER; John C. ;   et al.

Patent Application Summary

U.S. patent application number 14/873296 was filed with the patent office on 2016-02-11 for conditionally immortalized long-term stem cells and methods of making and using such cells. The applicant listed for this patent is National Jewish Health, THE REGENTS OF THE UNIVERSITY OF COLORADO. Invention is credited to John C. CAMBIER, Sara Ann JOHNSON, Yosef REFAELI, Brian Curtis Turner.

Application Number20160040129 14/873296
Document ID /
Family ID37963171
Filed Date2016-02-11

United States Patent Application 20160040129
Kind Code A1
CAMBIER; John C. ;   et al. February 11, 2016

CONDITIONALLY IMMORTALIZED LONG-TERM STEM CELLS AND METHODS OF MAKING AND USING SUCH CELLS

Abstract

Disclosed are methods for conditionally immortalizing stem cells, including adult and embryonic stem cells, the cells produced by such methods, therapeutic and laboratory or research methods of using such cells, and methods to identify compounds related to cell differentiation and development or to treat diseases, using such cells. A mouse model of acute myeloid leukemia (AML) and cells and methods related to such mouse model are also described.


Inventors: CAMBIER; John C.; (Denver, CO) ; REFAELI; Yosef; (Denver, CO) ; JOHNSON; Sara Ann; (Denver, CO) ; Turner; Brian Curtis; (Denver, CO)
Applicant:
Name City State Country Type

National Jewish Health
THE REGENTS OF THE UNIVERSITY OF COLORADO

Denver
Boulder

CO
CO

US
US
Family ID: 37963171
Appl. No.: 14/873296
Filed: October 2, 2015

Related U.S. Patent Documents

Application Number Filing Date Patent Number
14509870 Oct 8, 2014
14873296
11583970 Oct 18, 2006 8883507
14509870
60728131 Oct 18, 2005
60765993 Feb 6, 2006

Current U.S. Class: 435/366 ; 435/375; 530/350
Current CPC Class: A61P 37/06 20180101; C12N 2501/2303 20130101; C12N 2501/145 20130101; A61P 41/00 20180101; C12N 2510/04 20130101; C12N 5/0635 20130101; C12N 2501/105 20130101; C12N 5/0636 20130101; C12N 2501/2306 20130101; C12N 2501/606 20130101; A61P 7/06 20180101; A61P 35/00 20180101; C12N 2501/113 20130101; C12N 2740/16322 20130101; A61P 35/02 20180101; C12N 2501/48 20130101; C12N 2501/125 20130101; A61P 37/04 20180101; C07K 2319/50 20130101; A61P 37/00 20180101; C07K 14/4738 20130101; A61P 17/00 20180101; C12N 5/0647 20130101; C07K 14/005 20130101; C12N 5/0646 20130101; C07K 14/4747 20130101; C07K 14/82 20130101; C07K 2319/10 20130101; C12N 15/62 20130101
International Class: C12N 5/0789 20060101 C12N005/0789; C07K 14/82 20060101 C07K014/82; C07K 14/005 20060101 C07K014/005; C12N 5/0783 20060101 C12N005/0783; C12N 5/0781 20060101 C12N005/0781

Claims



1.-55. (canceled)

56. A fusion protein, comprising: a. a protein transduction domain of a Tat protein; and b. a MYC protein that is attached to the protein transduction domain of the Tat protein, wherein the fusion protein has MYC activity.

57. The fusion protein of claim 56, wherein the Tat protein is HIV-1 Tat.

58. The fusion protein of claim 56, wherein the MYC protein is capable of promoting in vitro cell proliferation, cell survival, or both.

59. The fusion protein of claim 56, wherein the MYC protein comprises the amino acid sequence of SEQ ID NO: 2.

60. A composition comprising: a. the fusion protein of claim 56; and b. a fusion protein comprising a Bcl-2 family member that inhibits apoptosis, and the protein transduction domain of a Tat protein.

61. The composition of claim 59, wherein the Bcl-2 family member is Bcl-2.

62. The composition of claim 61, wherein Bcl-2 comprises the amino acid sequence of SEQ ID NO: 6 or SEQ ID NO: 8.

63. A method for promoting one or more of cell proliferation or cell survival of one or more cells cultured in vitro comprising: providing to the one or more cells the fusion protein of claim 56; and culturing the one or more cells in a media.

64. The method of claim 63, wherein the culturing promotes in vitro cell proliferation, cell survival, or both; and wherein one or more of the survival or proliferation of the one or more cells is increased as compared with corresponding cells not exposed to the fusion protein or to the composition.

65. The method of claim 63, wherein the one or more cells are immune cells.

66. The method of claim 65, wherein the one or more immune cells are activated immune cells.

67. The method of claim 66, wherein the one or more immune cells are T cells.

68. The method of claim 66, wherein the one or more immune cells are B cells.

69. The method of claim 66, wherein the one or more immune cells are NK cells.

70. The method of claim 63, wherein the one or more cells are stem cells.

71. The method of claim 70, wherein the one or more stem cells are hematopoietic stem cells.

72. The method of claim 70, wherein the one or more stem cells are human stem cells.

73. The method of claim 70, wherein the one or more stem cells are genetically modified.

74. A method for promoting one or more of cell proliferation or cell survival of one or more cells cultured in vitro comprising: providing to the one or more cells the composition of claim 60; and culturing the one or more cells in a media.
Description



CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application is a continuation of U.S. application Ser. No. 14/509,870, filed Oct. 8, 2014, which is a continuation of U.S. application Ser. No. 11/583,970, filed Oct. 18, 2006, now U.S. Pat. No. 8,883,507, which claims the benefit of U.S. Provisional Patent Application No. 60/728,131, filed Oct. 18, 2005, and U.S. Provisional Application No. 60/765,993, filed Feb. 6, 2006. The entire disclosure of each of U.S. Provisional Patent Application No. 60/728,131 and U.S. Provisional Patent Application No. 60/765,993, U.S. application Ser. No. 14/509,870, and U.S. application Ser. No. 11/583,970 is incorporated herein by reference.

REFERENCE TO SEQUENCE LISTING

[0002] This application contains a Sequence Listing submitted on a compact disc, in duplicate. Each of the two compact discs, which are identical to each other pursuant to 37 CFR .sctn.1.52(e)(4), contains the following file: "2879-117.ST25.txt", having a size in bytes of 117 KB, recorded on 18 Oct. 2006. The information contained on the compact disc is hereby incorporated by reference in its entirety pursuant to 37 CFR .sctn.1.77(b)(4).

FIELD OF THE INVENTION

[0003] The present invention generally relates to conditionally immortalized long term stem cells, to methods of producing such cells, and to methods of using such cells, including therapeutic methods and drug discovery methods.

BACKGROUND OF THE INVENTION

[0004] The ability to manipulate the bone marrow output of various blood cells has become an important tool in the management of several diseases. Some of the best new therapies for hematological malignancies are based on the development of compounds that push leukemic cells to differentiate into lineages to which they are committed prior to the transforming event. One such example is the case of acute promyelocytic leukemia. Upon treatment of patients with Arsenic Trioxide, the malignant cells are pushed along the myelomonocytic pathway leading to remission of those tumors. Another example lies in promotion of successful engraftment of transplanted bone marrow stem cells (long term reconstituting hematopoietic stem cells, or lt-HSC) in irradiated individuals. The appearance of differentiated blood cells can be accelerated by the systemic administration of cytokines that are known to specifically induce red blood cell development (erythropoietin, or Epo), or myeloid cell development (granulocyte-macrophage colony-stimulating factor, or GM-CSF). Finally, harvesting of lt-HSC from donors has been greatly simplified by the process of "mobilization" wherein these cells are induced to move from the bone marrow sites where they normally reside into peripheral blood by systemic administration of a cytokine called G-CSF. Stem cells can then by harvested from peripheral blood obviating the painful and elaborate collection of bone marrow biopsies. All of these processes rely on the ability to program and control the biological behavior of lt-HSC.

[0005] Accordingly, bone marrow (stem cell) transplantation is an invaluable therapeutic tool for hematologic and immune reconstitution of individuals who have undergone radiation and/or chemotherapy (e.g. cancer patients, or have been exposed to high-level radiation), and is also a critical modality for treatment of immune deficiency and hematological malignancies. In addition, bone marrow transplantation would be a highly useful therapy to combat the negative effects of aging on the immune system, as well as on other cells and tissues. It is estimated that stem cell transplantation could benefit more than 35,000 children and adults per year.

[0006] The operative principle behind bone marrow transplantation is replacement of radiation sensitive lt-HSC that give rise to all blood cell types. Recent studies indicate that bone marrow transplantation may have value in the treatment of heart disease. Although the basis of this affect is unknown, it, and other findings, raise the possibility that hematopoietic stem cells (lt-HSC) may be reprogrammed to give rise to other tissues. If this is true, lt-HSC may have much broader utility and provide an alternative to controversial embryonic stem cell therapy.

[0007] The major obstacles confronting clinical application of bone marrow transplantation lie first in identification of an appropriately histocompatible marrow donor. This is usually accomplished using registries that have enrolled more than 6 million potential donors. The selected donor must undergo a grueling ordeal of induced mobilization stem cell into the blood followed by 4-5 days of leukapheresis to isolate rare lt-HSC. Transplantation of these cells must be followed by careful monitoring and treatment of the recipient to minimize graft versus host reactions caused by passenger lymphocytes.

[0008] Elucidation of the molecular basis of the impairment in hematopoietic lineage development has been complicated historically by the low frequency of relevant cell populations, which prevents biochemical analysis of signaling and downstream responses. In fact, this has been a major limiting factor in all studies of hematopoiesis. In addition, the limited availability of long-term hematopoietic stem cells (LT-HSCs) has also been a major obstacle in the treatment of many types of cancer as well as several kinds of immune deficiencies in humans. To the best of the present inventors' knowledge, there are currently no available cell lines that arose spontaneously that resemble lt-HSCs and can differentiate into normal lineages in vitro, or that can reconstitute lethally irradiated mice or sublethally irradiated humans, nor have any methods been described to deliberately generate such cell lines. Moreover, there are currently no viable technologies to continuously expand lt-HSCs, such that these cells need to be obtained from a donor every time they are needed.

[0009] There is also a dire need for additional modalities to treat hematological malignancies and immune deficiency, and novel cytokines to increase the output of transplanted lt-HSC. In addition, an appropriate platform for target identification and drug discovery does not currently exist. The missing elements are cell lines that represent different developmental stages in hematopoietic lineages. Optimally, such cells should retain the ability to undergo further differentiation in a specific lineage. Such cell lines are essential for identification of gene products, and thus new drugable targets, involved in regulation of cell development, proliferation and survival. In addition, such cell lines are essential for the screening of small molecule and shRNA libraries for loss-of function studies, as well as cDNA libraries for gain of function studies, in search of novel drugs.

[0010] Barriers to current drug discovery in this area include: (a) isolation of a sufficient number of cells from a particular developmental stage; (b) propagation of the cells in vitro for a sufficient length of time; and (c) ability to use conditional oncogenes to screen for drugs that could affect leukemic cells and not normal HSCs or progenitors.

[0011] Therefore, there is a great need in the art for a method to generate lt-HSC cell lines that can be expanded extensively, frozen, and used again whenever they are required, in the absence of subsequent harvests from the donor.

SUMMARY OF THE INVENTION

[0012] One embodiment of the present invention relates to a method to produce conditionally immortalized adult stem cells. The method includes the steps of: (a) obtaining an expanded population of adult stem cells; (b) transfecting the stem cells with a nucleic acid molecule comprising a protooncogene or biologically active fragment or homologue thereof that promotes cell survival and proliferation, wherein the protooncogene is inducible; (c) transfecting the stem cells with a nucleic acid molecule encoding a protein that inhibits apoptosis of the cell; and (d) expanding the transfected cells in the presence of a combination of stem cell growth factors under conditions whereby the protooncogene is active, to produce conditionally immortalized adult stem cells. In one aspect of this embodiment, the nucleic acid molecule of (b) and/or (c) is contained in an integrating vector. In one aspect, the nucleic acid molecule of (b) and/or (c) is transfected into the cells using a virus or viral vector selected from: retroviral vectors, lentivirus vectors, parvovirus, vaccinia virus, coronavirus, calicivirus, papilloma virus, flavivirus, orthomixovirus, togavirus, picornavirus, adenoviral vectors, modified and attenuated herpesviruses. In one aspect, the nucleic acid molecule of (b) and/or (c) is transfected into the cells using direct electroporation. In one aspect, the nucleic acid molecule or (b) and/or (c) is contained in a vector comprising a nucleic acid sequence encoding a drug-sensitivity protein. In one aspect, the nucleic acid molecule or (b) and/or (c) is contained in a vector comprising nucleic acid sequences encoding recognition substrate sequences for a recombinase flanking the nucleic acid molecule of (b) or (c).

[0013] In one aspect, this embodiment includes the additional steps of: (e) removing the conditions of (d) whereby the protooncogene is active; and (f) culturing the cells of (e) in media comprising growth factors that induce differentiation of the cells. This method can further include: (g) adding to the cells of (f), the conditions of (d) whereby the protooncogene is active, to produce conditionally immortalized cells in an intermediate stage of cell differentiation.

[0014] Another embodiment of the present invention relates to a method to produce conditionally immortalized adult stem cells, comprising: (a) obtaining an expanded population of adult stem cells; (b) culturing the stem cells in the presence of: (1) a combination of stem cell growth factors; (2) a first Tat-fusion protein, wherein Tat is fused to a protein encoded by a protooncogene or biologically active fragment or homologue thereof that promotes cell survival and proliferation; and (3) a second Tat-fusion protein, wherein Tat is fused to a protein that inhibits apoptosis in the stem cells.

[0015] Yet another embodiment of the present invention relates to method to produce conditionally immortalized embryonic stem cells, comprising: (a) obtaining an expanded population of embryonic stem cells; (b) transfecting the stem cells with a nucleic acid molecule comprising a protooncogene or biologically active fragment or homologue thereof that promotes cell survival and proliferation, wherein the protooncogene is inducible; (c) transfecting the stem cells with a nucleic acid molecule encoding a protein that inhibits apoptosis of the cell; and (d) expanding the transfected cells in the presence of a combination of stem cell growth factors under conditions whereby the protooncogene is active, to produce conditionally immortalized embryonic stem cells.

[0016] Another embodiment of the present invention relates to method to produce conditionally immortalized stem cells, comprising: (a) obtaining an expanded population of stem cells; (b) culturing the stem cells in the presence of: (1) a combination of stem cell growth factors; (2) a protein encoded by a protooncogene or biologically active fragment or homologue thereof that promotes cell survival and proliferation; and; (3) a protein that inhibits apoptosis in the stem cells. The protein of (2) and (3) are delivered into the stem cells using any suitable delivery system, including, but not limited to, Tat fusion, aptamers technology, or CHARIOT.TM. technology.

[0017] Yet another embodiment of the present invention relates to a method to produce conditionally immortalized stem cells, comprising: (a) obtaining an expanded population of stem cells; (b) delivering into the cells a protein encoded by a protooncogene or biologically active fragment or homologue thereof that promotes cell survival and proliferation, or a nucleic acid molecule encoding the same, wherein the protooncogene is inducible; (c) inhibiting apoptosis in the stem cells by delivering into the cells a protein that inhibits apoptosis of the cell, a nucleic acid molecule encoding the protein that inhibits apoptosis of the cell, or a nucleic acid molecule or protein that inhibits a proapoptotic protein in the cells; and (d) expanding the cells in the presence of a combination of stem cell growth factors under conditions whereby the protooncogene is active, to produce conditionally immortalized adult stem cells.

[0018] In any of the embodiments described above, the protooncogene can be selected from, but is not limited to: MYC-ER and ICN-1-ER. In any of the embodiments described above, the protein that inhibits apoptosis can be selected from, but is not limited to a member of the Bcl-2 family that inhibits apoptosis, such as Bcl-2, Bcl-X, Bcl-w, BclXL, Mcl-1, Dad-1, or hTERT. When the protooncogene is MYC-ER or ICN-1-ER, the conditions under which the protooncogene is active can include the presence of tamoxifen or an agonist thereof. In one aspect the cells are transfected with or are delivered (as a protein) MYC-ER and Bcl-2; MYC-ER and hTERT; ICN-1-ER and Bcl-2; ICN-1-ER and hTERT; or MYC-ER and ICN-1-ER.

[0019] In any of the embodiments described above, the step of expanding can be conducted in a medium including, but not limited to, (1) interleukin-6 (IL-6), IL-3 and stem cell factor (SCF); (2) a serum-free medium comprising stem cell factor (SCF), thrombopoietin (TPO), insulin-like Growth Factor 2 (IGF-2) and fibroblast Growth Factor 1 (FGF-1).

[0020] In any of the embodiments described above, the adult stem cells can include, but are not limited to: hematopoietic stem cells, intestinal stem cells, osteoblastic stem cells, mesenchymal stem cells, neural stem cells, epithelial stem cells, cardiac myocyte progenitor stem cells, skin stem cells, skeletal muscle stem cells, and liver stem cells. In one aspect, the mesenchymal stem cells are selected from lung mesenchymal stem cells and bone marrow stromal cells. In one aspect, the epithelial stem cells are selected from the group consisting of lung epithelial stem cells, breast epithelial stem cells, vascular epithelial stem cells and intestinal epithelial stem cells. In one aspect, the skin stem cells are selected from the group consisting of epidermal stem cells and follicular stem cells (hair follicle stem cells). In one aspect, the neural cells are selected from neuronal dopaminergic stem cells and motor-neuronal stem cells. In one aspect, the stem cells are from fresh or cryopreserved cord blood. In one aspect, the stem cells are hematopoietic progenitor cells obtained from the peripheral blood of normal or granulocyte colony-stimulating factor (G-CSF) treated patients.

[0021] In any of the embodiments described above, the method can further include genetically modifying the stem cells to correct a genetic defect in the cells, genetically modifying the stem cells to silence the expression of a gene, and/or genetically modifying the stem cells to overexpress a gene.

[0022] In any of the embodiments described above, the method can further include storing the cells. In one aspect, the method further includes retrieving the cells from storage and culturing the cells.

[0023] Another embodiment of the present invention relates to cells produced by any method described above or elsewhere herein.

[0024] Yet another embodiment of the present invention relates to a method to provide adult stem cells, or cells differentiated therefrom, to an individual comprising: (a) providing a source of conditionally immortalized adult stem cells produced by any method described above or elsewhere herein; (b) removing the conditions under which the stem cells of (a) are conditionally immortalized; and (c) administering the stem cells or cells differentiated therefrom to the individual. In one aspect, the cells were previously obtained from the individual in (c). In one aspect, the cells were obtained from a previously frozen stock of said cells. In one aspect, the cells are freshly obtained from the individual and conditionally immortalized by any method described above or elsewhere herein. In one aspect, the individual has cancer. In another aspect, the individual has leukemia. In another aspect, the individual has an immune deficiency disorder. In another aspect, the individual has an anemia disorder. In another aspect, the individual is undergoing reconstructive surgery. In another aspect, the individual is undergoing elective cosmetic surgery. In another aspect, the individual is undergoing transplantation surgery. In one aspect, the individual is in need of stem cells, or cells differentiated therefrom, selected from: hematopoietic stem cells, intestinal stem cells, osteoblastic stem cells, mesenchymal stem cells, neural stem cells, epithelial stem cells, cardiac myocyte progenitor stem cells, skin stem cells, skeletal muscle stem cells, and liver stem cells. In another aspect, the individual is in need of improved immune cell function. In another aspect, the individual has a genetic defect that is corrected by the stem cell.

[0025] Yet another embodiment of the present invention relates to a method to identify compounds that regulate lineage commitment and/or cell differentiation and development, comprising: (a) contacting adult stem cells produced by any method described above or elsewhere herein; and (b) detecting at least one genotypic or phenotypic characteristic in the stem cells of (a), as compared to the stem cells in the absence of the compound, wherein detection of a difference in the characteristic in the presence of the compound indicates that the compound affects the characteristic in the stem cell.

[0026] Another embodiment of the present invention relates to a method to study lineage commitment and/or cell differentiation and development, comprising evaluating adult stem cells produced by any method described above or elsewhere herein, or cells differentiated therefrom, to detect at least one genotypic or phenotypic characteristic of the cells.

[0027] Yet another embodiment of the present invention relates to the use of the cells produced by any method described above or elsewhere herein in a medicament for treating a condition or disease in which transplantation of stem cells is beneficial.

[0028] Another embodiment of the present invention relates to a mouse model of acute myeloid leukemia (AML), comprising a mouse produced by a method comprising: (a) lethally irradiating a mouse; (b) transferring conditionally immortalized long-term stem cells produced by any method described above or elsewhere herein and whole bone marrow cells from a Rag.sup.-/- mouse into the mouse; and (c) injecting periodic doses of tamoxifen or an agonist thereof into the mouse until the mouse develops clinical signs of AML. In one aspect, the cells are transfected with or are delivered (as a protein) MYC-ER and Bcl-2.

[0029] Another embodiment of the invention relates to tumor cells obtained from the mouse model of AML described above.

[0030] Yet another embodiment of the invention relates to the use of the mouse model of AML for preclinical testing of drug candidates specific for human proteins; to identify, develop, and/or test a compound for use in the diagnosis of, study of, or treatment of AML; or to identify, develop, and/or test a target for use in the diagnosis of, study of, or treatment of AML.

BRIEF DESCRIPTION OF THE DRAWINGS OF THE INVENTION

[0031] FIG. 1 is a graph showing mortality curves following bone marrow transplantation of transduced cells and activation of MYC function with 4OHT, in vivo.

[0032] FIG. 2 is a scatter plot showing scatter characteristics and GFP expression levels of HSCs derived from young and aged mice, following in vitro transduction. The dot plots represent the flow cytometric data for the forward (FSC) and side (SSC) scatter characteristics of the HSCs after three days in culture with IL-3, IL-6 and SCF. These two criteria correlate with cell size (FSC) and granularity (SSC).

[0033] FIGS. 3A and 3B are scatter plots showing the phenotypic comparison of cell lines derived from irradiated recipients reconstituted using BCL-2, MYC-ER and EGFP-transduced hematopoietic stem cells from aged (>60% ID.sup.- repertoire) and young 3-83.mu..delta. transgenic mice. Shown is the phenotype of representative clones 3 (young) and 3 (aged) months after initiation of culture.

[0034] FIG. 4 is a scatter plot showing the spontaneous differentiation of the aged LT-HSC line (ABM46) in vitro following withdrawal of tamoxifen (stem cell and B lineage marker expression are analyzed by flow cytometry).

[0035] FIGS. 5A and 5B are scatter plots showing the analysis of hematopoietic cell compartments derived from LT-HSC lines 6 weeks after adoptive transfer into irradiated young recipients. Data from three mice are presented in this figure, one mouse received the aged HSC line ABM42, and two mice received aged HSC line ABM46.

[0036] FIG. 6 is a scatter plot showing that the development of the B-cell compartment is compromised in mice reconstituted with ABM42 and ABM46 cell lines. Data from three mice are presented in this figure, one mouse received the aged HSC line ABM42, and two mice received aged HSC line ABM46.

[0037] FIG. 7 is a scatter plot showing T-cell development in mice that were reconstituted with ABM42 and ABM46 cell lines. Data from three mice are presented in this figure, one mouse received the aged HSC line ABM42, and two mice received aged HSC line ABM46.

[0038] FIG. 8 is a scatter plot and graph showing the phenotypic comparison of cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with BCL-2 and MYC-ER and maintained in continuous in vitro culture for >90 days. The panels represent the results of the flow cytometric analysis for expression of the viral expression markers (GFP and Thy1.1), as well as four markers required to define long-term HSCs in mice, Sca-1, c-kit, CD34 and Flk-2. The four cell lines contained subpopulations that retained the phenotypes of lt-HSCs (Sca-1+, c-kit+, CD34-, flk-2-).

[0039] FIG. 9 is a scatter plot and graph showing a phenotypic comparison of cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with different combinations of oncogenes and maintained in continuous in vitro culture for >90 days (pMIG-MYC and pMIT-Bcl-2 (top panels), pMIG-MYC.ER and pMIG-hTERT (middle panels), or pMIG-ICN.1.ER and pMIT-Bcl-2 (bottom panels)).

[0040] FIG. 10 is a scatter plot and graph showing a phenotypic comparison of cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with different combinations of oncogenes and maintained in continuous in vitro culture for >90 days (pMIG-ICN.1.ER and pMIT-Bcl-2 (top panels), pMIG-ICN.1 and pMIT-Bcl-2 (second row panels), or pMIG-ICN.1 and pMIG-Bcl-2 (third row panels), or pMIG-hTERT and pMIT-Bcl-2 (bottom panels)).

[0041] FIG. 11 is a scatter plot and graph showing a phenotypic comparison of cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with different combinations of oncogenes and maintained in continuous in vitro culture for >90 days (pMIG-MYC and pMIG-ICN.1 (top panels), pMIG-MYC.ER and pMIG-ICN.1 (middle panels), or pMIG-ICN.1.ER and pMIG-MYC (bottom panels).

[0042] FIG. 12 is a scatter plot showing the in vivo reconstitution of T cell and B cell compartments from cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with different combinations of oncogenes and maintained in continuous in vitro culture for >90 days.

[0043] FIG. 13 is a schematic drawing showing the use of recognition substrate sequences (RSS's) for recombinases in order to ensure the excision of recombinant DNA from conditionally immortalized long-term stem cells of the invention prior to transplantation.

[0044] FIG. 14 is a graph showing the detection of cells of the NK and erythroid lineage differentiated from conditionally immortalized long-term stem cells of the invention.

DETAILED DESCRIPTION OF THE INVENTION

[0045] The present invention provides a solution to the problem of being able to generate, maintain and manipulate stable cell lines derived from long-term stem cells, and particularly, long-term hematopoietic stem cells (lt-HSCs), that can give rise to all cell lineages that would normally arise from such cells when placed under the appropriate conditions. The present invention generally relates to methods to produce conditionally immortalized, long-term stem cells, to the stem cells produced by such methods, and to methods of using such stem cells. More specifically, using long-term hematopoietic stem cells as an exemplary stem cell population, the present inventors have established a powerful method to produce stem cells that are conditionally immortalized (e.g., reversibly immortalized or immortalized under specified conditions which is reversible when such conditions are removed), such stem cells being capable differentiating into normal cell lineages in vitro and in vivo, and being capable of reconstituting subjects in need of such cells. Indeed, the present invention can eliminate the need for a bone morrow donor, since the invention provides for the ability to harvest stem cells from a patient prior to a procedure (e.g. chemotherapy, radiation, etc.) to expand such cells, and return them to the patient. Moreover, such stem cells can be expanded extensively, stored (e.g., frozen), and then retrieved and expanded again, manipulated, and/or used repeatedly as required or desired. Such stem cells can be manipulated, for example, to correct a genetic defect or provide a benefit to a subject (therapeutic or preventative), or differentiated into a desired cell type. Finally, such cells can be used in a variety of assays for the identification of new targets involved in regulation of cell development, proliferation and survival, and the identification and development of drugs useful in ameliorating or treating diseases and conditions that would benefit from the regulation of cell development, proliferation and/or survival.

[0046] The present inventors have developed novel technology that allows the conditional immortalization of long-term stem cells, exemplified herein by long-term hematopoietic stem cells (lt-HSCs). The resulting cell lines can be expanded (propagated) indefinitely and exponentially in vitro and/or cryopreserved (stored), and have the ability to rescue lethally irradiated mice and to reconstitute all blood cell lineages in such animals. Furthermore, the inventors have been able to generate differentiated blood cells in vitro by extinguishing the function of the transforming oncogene. Such cells and the methods of producing them as described herein will allow the generation of transplantable human stem cells that carry no recombinant DNA, and thus pose no long term risk to the recipient. These conditionally immortalized lt-HSC's of the invention can be stabilized in their mature phenotypes and cell lines established in which the mature phenotype is preserved after reactivation of the oncogene. For example, the inventors have been able to develop CD4.sup.+ .alpha..beta..sup.+ T cells, as well as dendritic cell lines.

[0047] Applied in the clinical setting, this technology has the following advantages over bone marrow transplantation:

[0048] 1. Very few lt-HSC are needed to establish clones;

[0049] 2. Clones represent a renewable resource that can be stored indefinitely and accessed quickly;

[0050] 3. The cost of this therapy should be much less than conventional bone marrow transplantation;

[0051] 4. Use of lt-HSC clones should mitigate the threat of graft-versus-host disease, and associated costs.

[0052] 5. The technology can, at least in some cases, mitigate the need for a bone marrow donor.

[0053] In addition, the present invention provides for the use of the conditionally transformed long-term stem cells, such as the lt-HSC cells, to generate cells representing differentiated lineages (e.g., differentiated hematopoietic lineages, including intermediate stages of development of hematopoietic lineages). For example, in addition to countless therapeutic and preventative applications, these cell lines will allow the identification of novel compounds that can induce differentiation of malignant cells, arrest their growth, or induce apoptosis. These cells will also permit screening for novel cytokines and growth factors that direct the differentiation of stem cells in a particular pathway. Such cell lines simply do not exist and will be essential for drug discovery.

[0054] More specifically, in an effort to overcome the limitations in the art with regard to the provision and use of long term populations of adult-derived stem cells (although the invention is not limited to adult-derived stem cells, as discussed below), the present inventors have developed novel methods of producing of conditionally transformed cell lines representing early hematopoietic stem cell progenitors. In a specific, non-limiting example of the technology described and exemplified herein, the strategy involved the transfection (e.g., by retroviral transduction) of bone marrow stem cells from 5-fluorouracil (5-FU)-treated 3-83.mu..delta. mice. The inventors utilized the pMSCV bisistronic retroviral vector with inserts encoding Bcl-2 and green fluorescent protein (GFP) (as a reporter gene), and MYC-ER and GFP (again as a reporter gene). MYC was selected because of its ability to substitute for cytokine-derived survival and proliferative signals in lymphocytes. By restricting the target cell, the inventors hypothesized that stem cell tumors would form. Importantly, MYC-ER function is tamoxifen dependent in this setting, allowing for the termination of MYC function and transformation by withdrawing tamoxifen from the animal or cultures. In cells transduced with MYC-ER, the fusion protein is produced, but is retained in the cytoplasm until exposed to tamoxifen. Bcl-2 was selected because of its ability to inhibit apoptosis of cells that would normally occur as a result of exposure to the MYC signals and more particularly, when MYC is "inactivated" or removed by withdrawal of the tamoxifen from the cells. This novel combination of gene types (i.e., the invention is not limited to these specific genes, as discussed in more detail below) is partly responsible for the successful production of conditionally immortalized stem cells according to the present invention, and can readily be extended to other similar combinations of genes, as discussed in detail below.

[0055] Recipients of the transduced stem cells described above produced tumors (in the presence of 4OHT), and tumor cells from the bone marrow, spleen and lymph node were harvested and placed in culture with tamoxifen and a stem cell growth factor cocktail. The present inventors have discovered that, in the absence of an appropriate combination of stem cell growth factors, the stem cells produced by the present method will stop growing and die within a short period of time. Therefore, the use of a stem cell growth factor "cocktail" (i.e., combination of appropriate or suitable growth factors for stem cells) after transfection of the cells with the combination of genes discussed above is a second important aspect of the method of the present invention. This cocktail, while having the general characteristic of promoting and maintaining the growth of the stem cells, is not limited to a particular combination of growth factors, and parameters for selection of such factors are discussed in detail below.

[0056] The stem cells generated by the method of the present invention could be expanded in culture and were homogeneously positive for e.g. Sca1, positive for Endoglin and ckit, and negative for CD34, Flt3, B220, CD19 and mIgM, which are indicative of the phenotype of lt-HSC, which is well-characterized in the art. These cells could be frozen (cryopreserved, or stored), and then easily recovered and cultured after freezing. Importantly, the recovered cells were homogenous in phenotype and exhibited the phenotype of lt-HSC (e.g., again, uniformly GFP bright cells were positive for Sca1, Endoglin and c-kit, and negative for CD34, Flt3, B220, CD19 and mIgM). This phenotype corresponds perfectly with the published characteristics of long term repopulating pluripotent stem cells (Reya et al., 2003, Nature 423:409-14) that provide all long-term reconstitution in mice.

[0057] The inventors have further developed this method so that it can be performed completely in vitro (i.e., the initial procedure was conducted partly in vivo as described above). The inventors have also demonstrated that other combinations of genes having similar characteristics as those described above also result in the conditional immortalization of lt-HSCs. Furthermore, the cell lines can be differentiated in vitro into hematopoietic lineages by removing the tamoxifen and providing the appropriate growth factors, and will differentiate in vivo into all hematopoietic lineages in recipient animals in which tamoxifen is withheld. In addition, the cells can be differentiated into intermediate levels of development that have a stable phenotype and retain their ability to further differentiate along their committed pathway upon application or removal of the appropriate signal (described herein). Such cells are invaluable for various therapeutic applications. All of these experiments are described in detail below and in the Examples.

[0058] The methods and cell lines of the present invention provide a unique opportunity not only to study in detail the molecular, biochemical and cellular events that are associated with the commitment of adult stem cells toward various cell lineages and to study the differentiation and development of stem cells into various cell lineages, but also provide unique therapeutic and drug discovery tools.

[0059] For example, the stem cell lines of the present invention provide a unique source of expandable stem cells for use in a variety of transplantation, therapeutic and preventative strategies, including the treatment of cancer, and particularly, cancer that is treated by radiation. In current therapy for leukemia, for example, limited access to bone marrow donors and finite supplies of stem cells from such donors severely limit the options for reconstitution of a patient after radiation therapy. The present invention solves this problem by providing a means to generate a continuously expandable and renewable supply of autologous stem cells or histocompatible stem cells that can be stored and recovered as needed. Such technology could ultimately ablate the need for bone marrow donors altogether. In addition, a variety of immune deficiency disorders and anemia disorders (e.g., aplastic anemia or hemolytic anemia) will also benefit greatly from this technology, since the present invention provides the ability to repopulate hematopoietic cells of an individual as needed by the individual. Furthermore, the aging process is associated with several important changes in the hematopoietic compartment, including the increasing inability to mount a productive immune response, among others. Hematopoietic stem cells from aged mice have been shown to contain a higher level of mRNAs for DNA-repair problems. This may ultimately affect their ability to self-renew, undergo differentiation, undergo proliferation, and survive in response to bone marrow cytokines. Therefore, an aging individual can also benefit from the present invention in that a continuous supply of healthy hematopoietic cells can be provided to correct or ameliorate such deficiencies.

[0060] The technology of the present invention is not limited to bone marrow stem cells, but can be applied to virtually any type of stem cell, and can be extended beyond adult-derived cells to embryonic stem cells.

[0061] In one example, another application of the present invention relates to the generation of continuously expandable and renewable hair follicle stem cells. The development of conditionally immortalized stem cells from this lineage can be use in the context of reconstructive surgery for burn victims, for any individual that undergoes chemotherapy and/or radiation therapy resulting in the irreversible loss of hair growth, as well as patients following any surgical procedure affecting the skull. Furthermore, such cells could be used for elective procedures that involve the induction of hair growth in individuals affected by hereditary pattern baldness. Similarly, application of the present invention to stem cells of the skin will be invaluable for use in wound healing and treatment of burn victims, as well as plastic reconstructive surgery for trauma and other patients, as well as elective surgeries, including, but not limited to, cosmetic surgery. Such cells can be additionally genetically manipulated to correct inborn or acquired genetic defects in young and aged individuals. One of skill in the art will understand based on this disclosure that benefits can be derived from the use of the present invention on various other stem cell populations, including, but not limited to, stem cells derived from lung, breast, and intestinal epithelium and stem cells derived from neural and cardiac tissue, to name just a few. Other stem cell types are referenced elsewhere herein.

[0062] In addition, the present invention provides the unique opportunity for an individual to have access to expandable supplies of autologous stem cells and cells differentiated therefrom as needed throughout the life of the individual. For example, as the body ages, it is known that immune function and immune memory deteriorates. However, using the technology provided by the present invention, it will be possible to repopulate an individual with new, autologous stem cells that are capable of differentiation into all of the cells of the hematopoietic lineage, thus providing the aged individual with a "young" immune system. In addition, stem cells generated by the present method can be stored and used as part of therapeutic protocols during the lifetime of the individual, should they be needed (e.g., in the event the individual develops a cancer or immune deficiency disease or has another need for newly generated, autologous cells of virtually any type).

[0063] The present invention also provides unique opportunities for gene therapy. Specifically, genetic defects can now be corrected or beneficial gene modifications can be introduced into somatic cells by manipulating autologous stem cells obtained from an individual that have been conditionally immortalized and expanded using the method of the present invention. The stem cells can then be reintroduced into the individual from which they were obtained.

[0064] The stem cells produced by the method of the invention can also be used in a variety of drug discovery assays. Since one can now produce virtually unlimited supplies of homogeneous stem cells that can readily be stored, recovered, expanded and manipulated, such stem cells can be used as stem cells or differentiated into various cell lineages and used in assays to test various compounds for effects on cell differentiation, gene expression, and cell processes. The cells can be manipulated prior to contact with the compounds, such as by genetic manipulation. Stem cells from individuals with genetic defects can be evaluated in such assays in order to identify therapeutic compounds (e.g., cancer therapeutics) and evaluate gene replacement therapies. Indeed, the technology of the present invention provides an opportunity to target the cells of a specific individual to identify drug candidates and therapeutic candidates and strategies that are "tailored" to the cells of an individual. An example of such an assay is described in detail below.

[0065] With regard to research and discovery in the area of lineage commitment and cell differentiation and development, prior to the present invention, such studies were severely hampered by the lack of access to and the inability to generate sufficient numbers of the desired cell population to perform desired experiments. For example, in order to identify or screen for intermediates in the differentiation of a particular progenitor cell line, a sufficient number of cells must be obtained to provide meaningful and reproducible results. The progenitor cell line should also retain the ability to further differentiate in the lineage to which it has already committed, hence making these novel tools that do not currently exists, nor are there other descriptions of technology needed to generate those cells. Using technologies available at the time of the invention, this was not possible. The present invention solves the problem by providing expandable and essentially unlimited supplies of homogeneous stem cells that can be used in a variety of experiments. This technology will greatly enhance research capabilities in the area of cell differentiation and discovery.

[0066] As discussed above, the method for conditionally immortalizing lt-HSCs of the present invention can be adapted for additional stem cells derived from other tissues. For example, by adapting the gene delivery and growth factors, if needed, the present invention can be applied to a variety of different stem cells as described below. Such cells can also be expanded in vitro, and proceed to differentiate upon inactivation of the oncogenes, as described herein for hematopoietic stem cells. These cells can then be used for therapeutic applications that include tissue repair and tissue regeneration/engineering. Accordingly, the MYC-ER and Bcl-2 combination of genes, or any of the other combinations described herein, can be transfected by any method described herein or deemed suitable by one of skill in the art given this disclosure (including by a variety of viral-mediated methods), into cells including, but not limited to, mesenchymal stem cells (including, but not limited to, lung mesenchymal stem cells, bone marrow stromal cells), neural stem cells including, but not limited to, neuronal dopaminergic stem cells and motor-neuronal stem cells), epithelial stem cells (including, but not limited to, lung epithelial stem cells, breast epithelial stem cells, and intestinal epithelial stem cells), cardiac myocyte progenitor stem cells, skin stem cells (including, but not limited to, epidermal stem cells and follicular stem cells), skeletal muscle stem cells, endothelial stem cells (e.g., lung endothelial stem cells), and liver stem cells, to generate conditionally immortalized cell lines that can be expanded in vitro and proceed to differentiate upon inactivation of the oncogenes. In addition to the therapeutic potential of such cell lines, these lines can be further modified in vitro (or ex vivo) in order to correct inborn genetic defects, and used for studying the molecular basis of early lineage commitment and differentiation. While these cells may be a novel source of potentially relevant therapeutic targets, these cell lines will also be useful for the screening of small molecules that either prevent or induce differentiation, and for the identification of novel compounds and molecular targets for various therapies, including, but not limited to, cancer therapy and immune deficiency therapy.

GENERAL DEFINITIONS

[0067] In accordance with the present invention, reference to an isolated nucleic acid molecule herein is a nucleic acid molecule that has been removed from its natural milieu (i.e., that has been subject to human manipulation), its natural milieu being the genome or chromosome in which the nucleic acid molecule is found in nature. As such, "isolated" does not necessarily reflect the extent to which the nucleic acid molecule has been purified, but indicates that the molecule does not include an entire genome or an entire chromosome in which the nucleic acid molecule is found in nature. An isolated nucleic acid molecule can include a gene. An isolated nucleic acid molecule that includes a gene is not a fragment of a chromosome that includes such gene, but rather includes the coding region and regulatory regions associated with the gene, but no additional genes that are naturally found on the same chromosome. An isolated nucleic acid molecule can also include a specified nucleic acid sequence flanked by (i.e., at the 5' and/or the 3' end of the sequence) additional nucleic acids that do not normally flank the specified nucleic acid sequence in nature (i.e., heterologous sequences). Isolated nucleic acid molecule can include DNA, RNA (e.g., mRNA), or derivatives of either DNA or RNA (e.g., cDNA, siRNA, shRNA). Although the phrase "nucleic acid molecule" primarily refers to the physical nucleic acid molecule and the phrase "nucleic acid sequence" primarily refers to the sequence of nucleotides on the nucleic acid molecule, the two phrases can be used interchangeably, especially with respect to a nucleic acid molecule, or a nucleic acid sequence, being capable of encoding a protein or domain of a protein.

[0068] Preferably, an isolated nucleic acid molecule of the present invention is produced using recombinant DNA technology (e.g., polymerase chain reaction (PCR) amplification, cloning) or chemical synthesis. Isolated nucleic acid molecules include natural nucleic acid molecules and homologues thereof, including, but not limited to, natural allelic variants and modified nucleic acid molecules in which nucleotides have been inserted, deleted, substituted, and/or inverted in such a manner that such modifications provide the desired effect (e.g., provision of an inducible protooncogene, as described herein).

[0069] A nucleic acid molecule homologue can be produced using a number of methods known to those skilled in the art (see, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Labs Press (1989)). For example, nucleic acid molecules can be modified using a variety of techniques including, but not limited to, classic mutagenesis techniques and recombinant DNA techniques, such as site-directed mutagenesis, chemical treatment of a nucleic acid molecule to induce mutations, restriction enzyme cleavage of a nucleic acid fragment, ligation of nucleic acid fragments, PCR amplification and/or mutagenesis of selected regions of a nucleic acid sequence, synthesis of oligonucleotide mixtures and ligation of mixture groups to "build" a mixture of nucleic acid molecules and combinations thereof. Nucleic acid molecule homologues can be selected from a mixture of modified nucleic acids by screening for the function of the protein encoded by the nucleic acid and/or by hybridization with a wild-type gene.

[0070] The minimum size of a nucleic acid molecule or polynucleotide of the present invention is a size sufficient to encode a protein useful in the present invention, such as a protein encoded by a protooncogene or functional portion thereof (i.e., a portion that has the biological activity of the full-length protein and that is sufficient for use in the method of the invention), or an anti-apoptotic protein or a functional portion thereof (i.e., a portion that has the biological activity of the full-length protein and that is sufficient for use in the method of the invention). Other nucleic acid molecules that may be useful in the present invention can include nucleic acid molecules of a minimum size sufficient to form a probe or oligonucleotide primer that is capable of forming a stable hybrid with the complementary sequence of a nucleic acid molecule encoding the natural protein (e.g., under moderate, high or very high stringency conditions), which is typically at least 5 nucleotides in length, and preferably ranges from about 5 to about 50 or about 500 nucleotides or greater, including any length in between, in whole number increments (i.e., 5, 6, 7, 8, 9, 10, . . . 33, 34, . . . 256, 257, . . . 500). There is no limit, other than a practical limit, on the maximal size of a nucleic acid molecule of the present invention, in that the nucleic acid molecule can include a sequence or sequences sufficient to be useful in any of the embodiments of the invention described herein.

[0071] As used herein, stringent hybridization conditions refer to standard hybridization conditions under which nucleic acid molecules are used to identify similar nucleic acid molecules. Such standard conditions are disclosed, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Labs Press, 1989. Sambrook et al., ibid., is incorporated by reference herein in its entirety (see specifically, pages 9.31-9.62). In addition, formulae to calculate the appropriate hybridization and wash conditions to achieve hybridization permitting varying degrees of mismatch of nucleotides are disclosed, for example, in Meinkoth et al., 1984, Anal. Biochem. 138, 267-284; Meinkoth et al., ibid., is incorporated by reference herein in its entirety.

[0072] More particularly, moderate stringency hybridization and washing conditions, as referred to herein, refer to conditions which permit isolation of nucleic acid molecules having at least about 70% nucleic acid sequence identity with the nucleic acid molecule being used to probe in the hybridization reaction (i.e., conditions permitting about 30% or less mismatch of nucleotides). High stringency hybridization and washing conditions, as referred to herein, refer to conditions which permit isolation of nucleic acid molecules having at least about 80% nucleic acid sequence identity with the nucleic acid molecule being used to probe in the hybridization reaction (i.e., conditions permitting about 20% or less mismatch of nucleotides). Very high stringency hybridization and washing conditions, as referred to herein, refer to conditions which permit isolation of nucleic acid molecules having at least about 90% nucleic acid sequence identity with the nucleic acid molecule being used to probe in the hybridization reaction (i.e., conditions permitting about 10% or less mismatch of nucleotides). As discussed above, one of skill in the art can use the formulae in Meinkoth et al., ibid. to calculate the appropriate hybridization and wash conditions to achieve these particular levels of nucleotide mismatch. Such conditions will vary, depending on whether DNA:RNA or DNA:DNA hybrids are being formed. Calculated melting temperatures for DNA:DNA hybrids are 10.degree. C. less than for DNA:RNA hybrids. In particular embodiments, stringent hybridization conditions for DNA:DNA hybrids include hybridization at an ionic strength of 6.times.SSC (0.9 M Na.sup.+) at a temperature of between about 20.degree. C. and about 35.degree. C. (lower stringency), more preferably, between about 28.degree. C. and about 40.degree. C. (more stringent), and even more preferably, between about 35.degree. C. and about 45.degree. C. (even more stringent), with appropriate wash conditions. In particular embodiments, stringent hybridization conditions for DNA:RNA hybrids include hybridization at an ionic strength of 6.times.SSC (0.9 M Na.sup.+) at a temperature of between about 30.degree. C. and about 45.degree. C., more preferably, between about 38.degree. C. and about 50.degree. C., and even more preferably, between about 45.degree. C. and about 55.degree. C., with similarly stringent wash conditions. These values are based on calculations of a melting temperature for molecules larger than about 100 nucleotides, 0% formamide and a G+C content of about 40%. Alternatively, T.sub.m can be calculated empirically as set forth in Sambrook et al., supra, pages 9.31 to 9.62. In general, the wash conditions should be as stringent as possible, and should be appropriate for the chosen hybridization conditions. For example, hybridization conditions can include a combination of salt and temperature conditions that are approximately 20-25.degree. C. below the calculated T.sub.m of a particular hybrid, and wash conditions typically include a combination of salt and temperature conditions that are approximately 12-20.degree. C. below the calculated T.sub.m of the particular hybrid. One example of hybridization conditions suitable for use with DNA:DNA hybrids includes a 2-24 hour hybridization in 6.times.SSC (50% formamide) at about 42.degree. C., followed by washing steps that include one or more washes at room temperature in about 2.times.SSC, followed by additional washes at higher temperatures and lower ionic strength (e.g., at least one wash as about 37.degree. C. in about 0.1.times.-0.5.times.SSC, followed by at least one wash at about 68.degree. C. in about 0.1.times.-0.5.times.SSC).

[0073] In one embodiment of the present invention, any amino acid sequence described herein, including truncated forms (fragments or portions) and homologues of such sequences, can be produced with from at least one, and up to about 20, additional heterologous amino acids flanking each of the C- and/or N-terminal end of the given amino acid sequence. The resulting protein or polypeptide can be referred to as "consisting essentially of" a given amino acid sequence. According to the present invention, the heterologous amino acids are a sequence of amino acids that are not naturally found (i.e., not found in nature, in vivo) flanking the given amino acid sequence or which would not be encoded by the nucleotides that flank the naturally occurring nucleic acid sequence encoding the given amino acid sequence as it occurs in the gene, if such nucleotides in the naturally occurring sequence were translated using standard codon usage for the organism from which the given amino acid sequence is derived. Similarly, the phrase "consisting essentially of", when used with reference to a nucleic acid sequence herein, refers to a nucleic acid sequence encoding a given amino acid sequence that can be flanked by from at least one, and up to as many as about 60, additional heterologous nucleotides at each of the 5' and/or the 3' end of the nucleic acid sequence encoding the given amino acid sequence. The heterologous nucleotides are not naturally found (i.e., not found in nature, in vivo) flanking the nucleic acid sequence encoding the given amino acid sequence as it occurs in the natural gene.

[0074] According to the present invention, a recombinant vector (also referred to generally as a recombinant nucleic acid molecule, particularly when it contains a nucleic acid sequence of interest according to the invention) is an engineered (i.e., artificially produced) nucleic acid molecule that is used as a tool for manipulating a nucleic acid sequence of choice and for introducing such a nucleic acid sequence into a host cell. The recombinant vector is therefore suitable for use in cloning, sequencing, and/or otherwise manipulating the nucleic acid sequence of choice, such as by expressing and/or delivering the nucleic acid sequence of choice into a host cell. Such a vector typically contains heterologous nucleic acid sequences, i.e., nucleic acid sequences that are not naturally or usually found adjacent to a nucleic acid sequence to be cloned or delivered, although the vector can also contain regulatory nucleic acid sequences (e.g., promoters, untranslated regions) which are naturally found adjacent to nucleic acid molecules of the present invention, or which are useful for expression of the nucleic acid molecules of the present invention (discussed in detail below). A vector can be either RNA or DNA, either prokaryotic or eukaryotic, and typically is a plasmid or a viral vector. The vector can be maintained as an extrachromosomal element (e.g., a plasmid) or it can be integrated into the chromosome of a host cell. The entire vector can remain in place within a host cell, or under certain conditions, the plasmid DNA can be deleted, leaving behind the nucleic acid molecule of the present invention. Under other conditions, the vector is designed to be excised (removed) from the genome of the host cell at a selected time (described in more detail below). The integrated nucleic acid molecule can be under chromosomal promoter control, under native or plasmid promoter control, or under a combination of several promoter controls. Single or multiple copies of the nucleic acid molecule can be integrated into the chromosome. A recombinant vector of the present invention can contain at least one selectable marker.

[0075] According to the present invention, the phrase "operatively linked" refers to linking a nucleic acid molecule to an expression control sequence (e.g., a transcription control sequence and/or a translation control sequence) in a manner such that the molecule can be expressed when transfected (i.e., transformed, transduced, transfected, conjugated or conduced) into a host cell. Transcription control sequences are sequences that control the initiation, elongation, or termination of transcription. Particularly important transcription control sequences are those that control transcription initiation, such as promoter, enhancer, operator and repressor sequences. Suitable transcription control sequences include any transcription control sequence that can function in a host cell or organism into which the recombinant nucleic acid molecule is to be introduced.

[0076] According to the present invention, the term "transfection" is used to refer to any method by which an exogenous nucleic acid molecule (i.e., a recombinant nucleic acid molecule) can be inserted into a cell. The term "transduction" is a specific type of transfection in which genetic material is transferred from one source to another, such as by a virus (e.g., a retrovirus) or a transducing bacteriophage. The term "transformation" can be used interchangeably with the term "transfection" when such term is used to refer to the introduction of nucleic acid molecules into microbial cells, such as bacteria and yeast. In microbial systems, the term "transformation" is used to describe an inherited change due to the acquisition of exogenous nucleic acids by the microorganism and is essentially synonymous with the term "transfection." However, in animal cells, transformation has acquired a second meaning that can refer to changes in the growth properties of cells in culture after they become cancerous, for example. Therefore, to avoid confusion, the term "transfection" is preferably used herein with regard to the introduction of exogenous nucleic acids into animal cells. Therefore, the term "transfection" will be used herein to generally encompass transfection or transduction of animal cells, and transformation or transduction of microbial cells, to the extent that the terms pertain to the introduction of exogenous nucleic acids into a cell. Transfection techniques include, but are not limited to, transformation, transduction, particle bombardment, diffusion, active transport, bath sonication, electroporation, microinjection, lipofection, adsorption, infection and protoplast fusion.

[0077] As used herein, reference to an isolated protein or polypeptide in the present invention includes full-length proteins, fusion proteins, chimeric proteins, or any fragment (truncated form, portion) or homologue of such a protein. More specifically, an isolated protein according to the present invention, is a protein (including a polypeptide or peptide) that has been removed from its natural milieu (i.e., that has been subject to human manipulation), and can include, but is not limited to, purified proteins, partially purified proteins, recombinantly produced proteins, membrane bound proteins, proteins complexed with lipids, soluble proteins, synthetically produced proteins, and isolated proteins associated with other proteins. As such, "isolated" does not reflect the extent to which the protein has been purified. Preferably, an isolated protein of the present invention is produced recombinantly. In addition, and again by way of example with respect to the naming of a particular protein (Bcl-2), a "human Bcl-2 protein" or a protein "derived from" a human Bcl-2 protein refers to a Bcl-2 protein (including a homologue or portion of a naturally occurring Bcl-2 protein) from a human (Homo sapiens) or to a Bcl-2 protein that has been otherwise produced from the knowledge of the structure (e.g., sequence) and perhaps the function of a naturally occurring Bcl-2 protein from Homo sapiens. In other words, a human Bcl-2 protein includes any Bcl-2 protein that has substantially similar structure and function of a naturally occurring Bcl-2 protein from Homo sapiens or that is a biologically active (i.e., has biological activity) homologue of a naturally occurring Bcl-2 protein from Homo sapiens as described in detail herein. As such, a human Bcl-2 protein can include purified, partially purified, recombinant, mutated/modified and synthetic proteins. According to the present invention, the terms "modification" and "mutation" can be used interchangeably, particularly with regard to the modifications/mutations to the amino acid sequence of a protein (or nucleic acid sequences) described herein.

[0078] As used herein, the term "homologue" is used to refer to a protein or peptide which differs from a naturally occurring protein or peptide (i.e., the "prototype" or "wild-type" protein) by modifications, including minor modifications, to the naturally occurring protein or peptide, but which maintains the basic protein and side chain structure of the naturally occurring form. Such changes include, but are not limited to: changes in one or a few (i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10) amino acid side chains; changes one or a few amino acids, including deletions (e.g., a protein or truncated form of the protein or peptide), insertions and/or substitutions; changes in stereochemistry of one or a few atoms; and/or minor derivatizations, including but not limited to: methylation, glycosylation, phosphorylation, acetylation, myristoylation, prenylation, palmitation, amidation and/or addition of glycosylphosphatidyl inositol. A homologue can have either enhanced, decreased, or substantially similar properties as compared to the naturally occurring protein or peptide. A homologue can include an agonist of a protein or an antagonist of a protein.

[0079] Homologues can be the result of natural allelic variation or natural mutation. A naturally occurring allelic variant of a nucleic acid encoding a protein is a gene that occurs at essentially the same locus (or loci) in the genome as the gene which encodes such protein, but which, due to natural variations caused by, for example, mutation or recombination, has a similar but not identical sequence. Allelic variants typically encode proteins having similar activity to that of the protein encoded by the gene to which they are being compared. One class of allelic variants can encode the same protein but have different nucleic acid sequences due to the degeneracy of the genetic code. Allelic variants can also comprise alterations in the 5' or 3' untranslated regions of the gene (e.g., in regulatory control regions). Allelic variants are well known to those skilled in the art.

[0080] Homologues can be produced using techniques known in the art for the production of proteins including, but not limited to, direct modifications to the isolated, naturally occurring protein, direct protein synthesis, or modifications to the nucleic acid sequence encoding the protein using, for example, classic or recombinant DNA techniques to effect random or targeted mutagenesis.

[0081] In one embodiment, a homologue of a given protein comprises, consists essentially of, or consists of, an amino acid sequence that is at least about 45%, or at least about 50%, or at least about 55%, or at least about 60%, or at least about 65%, or at least about 70%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95% identical, or at least about 95% identical, or at least about 96% identical, or at least about 97% identical, or at least about 98% identical, or at least about 99% identical (or any percent identity between 45% and 99%, in whole integer increments), to the amino acid sequence of the reference protein. In one embodiment, the homologue comprises, consists essentially of, or consists of, an amino acid sequence that is less than 100% identical, less than about 99% identical, less than about 98% identical, less than about 97% identical, less than about 96% identical, less than about 95% identical, and so on, in increments of 1%, to less than about 70% identical to the naturally occurring amino acid sequence of the reference protein.

[0082] As used herein, unless otherwise specified, reference to a percent (%) identity refers to an evaluation of homology which is performed using: (1) a BLAST 2.0 Basic BLAST homology search using blastp for amino acid searches and blastn for nucleic acid searches with standard default parameters, wherein the query sequence is filtered for low complexity regions by default (described in Altschul, S. F., Madden, T. L., Schaaffer, A. A., Zhang, J., Zhang, Z., Miller, W. & Lipman, D. J. (1997) "Gapped BLAST and PSI-BLAST: a new generation of protein database search programs." Nucleic Acids Res. 25:3389-3402, incorporated herein by reference in its entirety); (2) a BLAST 2 alignment (using the parameters described below); (3) and/or PSI-BLAST with the standard default parameters (Position-Specific Iterated BLAST. It is noted that due to some differences in the standard parameters between BLAST 2.0 Basic BLAST and BLAST 2, two specific sequences might be recognized as having significant homology using the BLAST 2 program, whereas a search performed in BLAST 2.0 Basic BLAST using one of the sequences as the query sequence may not identify the second sequence in the top matches. In addition, PSI-BLAST provides an automated, easy-to-use version of a "profile" search, which is a sensitive way to look for sequence homologues. The program first performs a gapped BLAST database search. The PSI-BLAST program uses the information from any significant alignments returned to construct a position-specific score matrix, which replaces the query sequence for the next round of database searching. Therefore, it is to be understood that percent identity can be determined by using any one of these programs.

[0083] Two specific sequences can be aligned to one another using BLAST 2 sequence as described in Tatusova and Madden, (1999), "Blast 2 sequences--a new tool for comparing protein and nucleotide sequences", FEMS Microbiol Lett. 174:247-250, incorporated herein by reference in its entirety. BLAST 2 sequence alignment is performed in blastp or blastn using the BLAST 2.0 algorithm to perform a Gapped BLAST search (BLAST 2.0) between the two sequences allowing for the introduction of gaps (deletions and insertions) in the resulting alignment. For purposes of clarity herein, a BLAST 2 sequence alignment is performed using the standard default parameters as follows.

[0084] For blastn, using 0 BLOSUM62 matrix:

[0085] Reward for match=1

[0086] Penalty for mismatch=-2

[0087] Open gap (5) and extension gap (2) penalties

[0088] gap x_dropoff (50) expect (10) word size (11) filter (on)

[0089] For blastp, using 0 BLOSUM62 matrix:

[0090] Open gap (11) and extension gap (1) penalties

[0091] gap x_dropoff (50) expect (10) word size (3) filter (on).

[0092] According to the present invention, an isolated protein, including a biologically active homologue or fragment thereof, has at least one characteristic of biological activity of activity the wild-type, or natural occurring protein. In general, the biological activity or biological action of a protein refers to any function(s) exhibited or performed by the protein that is ascribed to the naturally occurring form of the protein as measured or observed in vivo (i.e., in the natural physiological environment of the protein) or in vitro (i.e., under laboratory conditions). Modifications, activities or interactions which result in a decrease in protein expression or a decrease in the activity of the protein, can be referred to as inactivation (complete or partial), down-regulation, reduced action, or decreased action or activity of a protein. Similarly, modifications, activities or interactions that result in an increase in protein expression or an increase in the activity of the protein, can be referred to as amplification, overproduction, activation, enhancement, up-regulation or increased action of a protein.

Method of Conditional Immortalization of the Invention

[0093] One embodiment of the present invention relates to a method to produce conditionally immortalized, adult stem cells, and preferably long-term stem cells. The method generally includes the following steps: (a) obtaining an expanded population of adult stem cells; (b) transfecting (transducing) the stem cells with a vector comprising a protooncogene that promotes cell survival and proliferation, wherein the protooncogene is regulatable (inducible, controllable), (c) transfecting (transducing) the stem cells with a vector encoding a protein that inhibits apoptosis of the cell; and (d) expanding the transfected cells in the presence of a combination of stem cell growth factors under conditions whereby the protooncogene is active. In one embodiment, the vector is an integrating vector. Cells produced by this method can be cultured, expanded, stored, recovered, used in therapeutic methods, used in research and discovery methods, genetically manipulated, induced to differentiate by removing the conditions whereby the protooncogene is active, and/or used in any other method described herein or apparent to one of skill in the art given this disclosure. Steps (b) and (c) can be performed in any order.

[0094] According to the present invention, the phrase "conditionally immortalized" refers to cells that are immortalized (e.g., capable of indefinite growth without differentiation in a cytokine dependent fashion, while maintaining their ability and potential to differentiate into a number of different lineages under the appropriate conditions) in a reversible manner, such that the cells are immortalized under a specific set of conditions, and when the conditions are removed or changed (or other conditions added), the cells are no longer immortalized and may differentiate into other cell types. The phrase "conditionally immortalized" can be used interchangeably with the phrase "reversibly immortalized". For example, referring to the method of the present invention, the presence of the regulatable protooncogene that promotes cell survival and proliferation causes the cells to retain an immortalized phenotype when the stem cell is placed under conditions that allow the protooncogene to be activated (e.g., tamoxifen or an agonist thereof in the case of MYC-ER). In other words, the cells grow and expand indefinitely in culture, and are maintained in an undifferentiated state under these specific conditions. When these conditions are removed (e.g., the tamoxifen is removed with respect to MYC-ER), the stem cells are no longer immortalized and can differentiate into various cell lineages given the appropriate environment (e.g., the appropriate combination of growth factors).

[0095] Reference to "stem cells", as used herein, refers to the term as it is generally understood in the art. For example, stem cells, regardless of their source, are cells that are capable of dividing and renewing themselves for long periods, are unspecialized (undifferentiated), and can give rise to (differentiate into) specialized cell types (i.e., they are progenitor or precursor cells for a variety of different, specialized cell types). "Long-term", when used in connection with stem cells, refers to the ability of stem cells to renew themselves by dividing into the same non-specialized cell type over long periods (e.g., many months, such as at least 3 months, to years) depending on the specific type of stem cell. As discussed herein, phenotypic characteristics of various long-term stem cells from different animal species, such as long-term hematopoietic stem cells (lt-HSC) are known in the art. For example, murine lt-HSC can be identified by the presence of the following cell surface marker phenotype: c-kit+, Sca-1+, CD34-, flk2- (see Examples). Adult stem cells include stem cells that can be obtained from any non-embryonic tissue or source, and typically generate the cell types of the tissue in which they reside. The term "adult stem cell" may be used interchangeably with the term "somatic stem cell". Embryonic stem cells are stem cells obtained from any embryonic tissue or source.

[0096] In one embodiment of the invention, the stem cells used in the present invention can include any adult stem cells obtained from any source. In another embodiment of the invention, stem cells can include embryonic stem cells. Stem cells useful in the present invention include, but are not limited to, hematopoietic stem cells, mesenchymal stem cells (including, but not limited to, lung mesenchymal stem cells, bone marrow stromal cells), neural stem cells, epithelial stem cells (including, but not limited to, lung epithelial stem cells, breast epithelial stem cells, vascular epithelial stem cells, and intestinal epithelial stem cells), intestinal stem cells, cardiac myocyte progenitor stem cells, skin stem cells (including, but not limited to, epidermal stem cells and follicular stem cells (hair follicle stem cells)), skeletal muscle stem cells, osteoblastic precursor stem cells, and liver stem cells.

[0097] Hematopoietic stem cells give rise to all of the types of blood cells, including but not limited to, red blood cells (erythrocytes), B lymphocytes, T lymphocytes, natural killer cells, neutrophils, basophils, eosinophils, monocytes, macrophages, and platelets.

[0098] Mesenchymal stem cells (including bone marrow stromal cells) give rise to a variety of cell types, including, but not limited to bone cells (osteocytes), cartilage cells (chondrocytes), fat cells (adipocytes), lung cells, and other kinds of connective tissue cells such as those in tendons.

[0099] Neural stem cells in the brain give rise to its three major cell types: nerve cells (neurons) and two categories of non-neuronal cells, astrocytes and oligodendrocytes.

[0100] Epithelial stem cells in the lining of various tissues give rise to several cell types that form the epithelium in tissues.

[0101] Skin stem cells occur in the basal layer of the epidermis and at the base of hair follicles. The epidermal stem cells give rise to keratinocytes, which migrate to the surface of the skin and form a protective layer, and the follicular stem cells can give rise to both the hair follicle and to the epidermis. Other sources of adult stem cells will be known to those of skill in the art.

[0102] Embryonic stem cells can give rise to all tissues and cells of the body.

[0103] Methods for obtaining such stem cells and providing initial culture conditions, such as a liquid culture or semi-solid culture medium, are known in the art. The cells are initially expanded in vivo or in vitro, by contacting the source of the stem cells with a suitable reagent that expands or enriches such cells in the tissue source or in culture. For example, in the case of hematopoietic stem cells, the donor individual can be treated with an agent that enriches for hematopoietic stem cells and encourages such cells to proliferate without differentiation, such as 5-fluorouracil. Other suitable agents for expansion of a desired stem cell type will be known to those of skill in the art. Alternatively, and preferably, adult stem cells are isolated from a tissue source and then expanded or enriched in vitro by exposure to a suitable agent. For example, with regard to hematopoietic stem cells, a method for producing an expanded culture of adult hematopoietic progenitors is described in Van Parijs et al., (1999; Immunity, 11, 763-70). Cells are obtained from an individual by any suitable method for obtaining a cell sample from an animal, including, but not limited, to, collection of bone marrow collection of a bodily fluid (e.g., blood), collection of umbilical cord blood, tissue punch, and tissue dissection, including particularly, but not limited to, any biopsies of skin, intestine, cornea, spinal cord, brain tissue, scalp, stomach, breast, lung (e.g., including lavage and bronchioschopy), fine needle aspirates of the bone marrow, amniotic fluid, placenta and yolk sac.

[0104] In one embodiment, cells useful in the invention can also be obtained from fresh, or cryopreserved (stored) cord blood, hematopoietic progenitor populations that can be derived from the directed differentiation of embryonic stem (ES) cells in vitro, hematopoietic stem cells (HSCs) obtained from the peripheral blood of normal or granulocyte colony-stimulating factor (G-CSF)-treated patients who have been induced to mobilize their lt-HSCs to the peripheral circulation.

[0105] Once an expanded population of stem cells is obtained (made available, provided, or produced), the cells are transfected, either sequentially (in any order) or simultaneously, with: (1) a vector comprising a protooncogene that promotes cell survival and proliferation, wherein the protooncogene is regulatable (inducible, controllable), and (2) a vector encoding a protein that inhibits apoptosis of the cell. Preferably, the vector is an integrating vector, defined herein as any vector that has the ability to integrate into the genome of a cell (e.g., a retroviral vector). Various vectors and methods of transfection are described in detail below. The protooncogene is regulatable (inducible or controllable), so that the protooncogene can be activated and deactivated (i.e., turned on or turned off) as desired to either maintain the stem cell in an immortalized state or to allow it to differentiate into a desired cell type. Protooncongenes can be selected, or designed, to be regulated by any suitable method, including in response to any condition, such as the presence or absence of a compound or agent, temperature, or any other suitable condition. By way of example, the protooncogenes MYC-ER (the estrogen receptor (ER)-regulated MYC) and ICN-1-ER (the ER-regulated intracellular portion of Notch-1) described herein are both inducible in the presence of tamoxifen. It is noted that such genes can also be engineered to be responsive to other dimerizing drugs, such as FK1012, altered forms of Rapamycin, or could be expressed from vectors that contain a tetracycline responsive element. The latter scenario regulates expression of the protein, not the function of a polypeptide present in the cell. Other similar modifications of this platform technology will be apparent to those of skill in the art.

[0106] The protooncogene useful in the method of the present invention is any protooncogene that promotes cell survival and proliferation. Preferred protooncogenes to use in the method of the invention include, but are not limited to MYC, ICN-1, hTERT, (reverse transcriptase component of the human telomerase), NMYC, S-MYC, L-MYC, Akt (myrystylated). In addition, other suitable genes to use or methods of the invention or ways to modify genes to achieve the desired result include, but are not limited to use of downstream signaling effectors such as pyruvate dehydrogenase kinase 1 (PDK-1); mammalian target of Rapamycin (mTOR); loss of phosphatase and tensin homologue (PTEN) by shRNA; Bcl-3, Cyclin D1, Cyclin D3, Bcl-10, Bcl-6, BCR-ABL (breakpoint cluster region fusion with ABL) and its various mutant forms, constitutively active forms of Stat5 and Stat3, AML1-ETO (fusion of acute myelogenous leukemia 1 and runt-related transcription factor 1), MLL-ENL (mixed lineage leukemia and eleven nineteen leukemia), Hox genes, activated forms of the interleukin-3 (IL-3) receptor .beta. chain, and other cytokine receptor chains (epidermal growth factor receptor (EGFR), c-kit, platelet-derived growth factor receptor (PDGFR), etc.), as well as wnt (all mammalian forms), .beta.-catenin, sonic hedgehog (shh-1 and all mammalian forms), bmi-1 and c-jun (all mammalian forms). Also, the present invention includes inducing the loss (or inhibition) of cyclin kinase inhibitors by shRNA, including, but not limited to, p16, p19, p21 and p27. In one embodiment, the present invention includes the use of regulatable homologues of any or such protooncogenes (e.g., MYC-ER or ICN-1-ER) or other genes. The Examples describe the use of both MYC-ER or ICN-1-ER to successfully produce conditionally immortalized lt-HSC using the method of present invention.

[0107] The nucleic acid sequence encoding human MYC is represented herein as SEQ ID NO:1, which encodes an amino acid sequence represented herein as SEQ ID NO:2. The nucleic acid sequence encoding hTERT is represented herein as SEQ ID NO:3, which encodes an amino acid sequence represented herein as SEQ ID NO:4. The nucleic acid sequence encoding human ICN-1 is represented herein as SEQ ID NO:11, which encodes an amino acid sequence represented herein as SEQ ID NO:12. ICN-1 a portion of Notch-1, and specifically, amino acids 1757-2555 from Notch-1 (see Aster et al., Mol Cell Biol. 2000 October; 20(20):7505-15, incorporated herein by reference in its entirety). The nucleotide and amino acid sequence for MYC-ER are known in the art and the MYC-ER protein is described in Soloman et al., Oncogene. 1995 Nov. 2; 11(9):1893-7, incorporated herein by reference in its entirety. ICN-1-ER was created by the present inventors and the nucleic acid sequence encoding this protein is represented herein as SEQ ID NO:13, which encodes an amino acid sequence represented by SEQ ID NO:14.

[0108] Similarly, a preferred anti-apoptosis gene is Bcl-2, although other genes that encode proteins that inhibit apoptosis and particularly, maintain cell survival when the protooncogene is inactivated in the stem cell, are included in the present invention. The nucleic acid sequence encoding Bcl-2 alpha is represented herein as SEQ ID NO:5, which encodes an amino acid sequence of SEQ ID NO:6. Bcl-2 beta is represented herein as SEQ ID NO:7, which encodes an amino acid sequence of SEQ ID NO:8. An "anti-apoptosis" gene is defined herein as any gene that encodes a protein that can inhibit (reduce, prevent, decrease) a process associated with apoptosis in a cell or promote (enhance, increase, stimulate, allow) cell survival, even in the presence of conditions that could induce apoptosis. Proteins associated with apoptosis, and the genes encoding such proteins, are well-known in the art. Such other genes include, but are not limited to, any genes in the Bcl-2 family that will likely be important in the setting of conditional transformation of adult stem cells (i.e., not just hematopoietic stem cells). These genes include, but are not limited to, other pro-survival members of the Bcl-2 family, such as Bcl-X, Bcl-w, BclXL, Mcl-1, Dad-1, or hTERT (reverse transcriptase component of the human telomerase, which has been shown to inhibit proliferation). Such genes are ectopically overexpressed in the presence of the regulated oncogene, as described with Bcl-2 in the working examples herein. In addition, this aspect of the present invention includes using shRNA mediated gene knockdown (or disruption or inhibition by any other method) for BH3-only members of the bcl-2 family that are proapoptotic (e.g., Bim, PUMA, NOXA, Bax, Bak, BclXS, Bad, Bar, and others), as well as disruption of Caspases 3, 9, 10, MLL-1 (and all mammalian forms), Enl-1 (Endospermless-1) and all mammalian forms, Apaf-1 and other elements that form part of the apoptosome.

[0109] The nucleic acid sequence for each of these genes described above or the coding region thereof is known in the art and is publicly available, including for humans. Similarly, the amino acid sequence for proteins encoded by these genes is known in the art and is publicly available.

[0110] The present inventors have produced several different long-term, conditionally immortalized stem cells using the method of the present invention and using different combinations of protooncogenes and anti-apoptotic genes, including the following combinations: MYC-ER and Bcl-2; MYC-ER and hTERT (reverse transcriptase component of the human telomerase); ICN-1-ER and Bcl-2; ICN-1-ER and hTERT; and MYC-ER and ICN-1-ER.

[0111] It is noted that with regard to either of the protooncogene or the gene encoding an anti-apoptosis protein used in the present method, it is not required that the entire gene be used in the constructs described herein, since any portion of the gene or a nucleic acid sequence (e.g., cDNA) that encodes the desired functional protein product, a functional portion thereof, or a functional homologue thereof is encompassed by the invention. Accordingly, reference generally herein to the genes or transgenes used to transfect stem cells is to be understood to be exemplary and to include the use of any nucleic acid molecules encoding the entire gene, the entire coding region of the gene, or portions of the genes or homologues thereof, as long as such nucleic acid sequences encode functional proteins suitable for use in the present invention.

[0112] In one embodiment of the present invention, the present method additional includes the use of shRNAs or siRNAs that are directed against RNAs encoding proapoptotic proteins, such as the pro-apoptotic members of the Bcl-2 family, namely those of the BH3-only type (Bim, Bax, Bak, Puma, Noxa, etc.). The disruption of a pro-apoptotic gene in the context of a regulated oncogene is expected to result in a more efficient immortalization of certain stem cell populations. RNA interference (RNAi) is a process whereby double stranded RNA, and in mammalian systems, short interfering RNA (siRNA) or short hairpin RNA (shRNA), is used to inhibit or silence expression of complementary genes. In the target cell, siRNA are unwound and associate with an RNA induced silencing complex (RISC), which is then guided to the mRNA sequences that are complementary to the siRNA, whereby the RISC cleaves the mRNA. shRNA is transfected into a target cell in a vector where it is transcribed, and then processed by DICER enzymes to form siRNA-like molecules that activate RISC, which, as with siRNA, is then guided to the mRNA sequences that are complementary to the shRNA, whereby the RISC cleaves the mRNA.

[0113] The stem cells can be transfected with the vectors comprising the protooncogene and encoding the anti-apoptosis protein using any suitable method of transfecting cells, and particularly mammalian cells, including by using combinations of techniques. The present inventors have discovered that it is the particular coordination between the genes (or constructs) that are expressed that have resulted in the generation of conditionally immortalized, long term stem cells as described herein. The Examples have demonstrated the use of retroviral vectors, but other methods include, but are not limited to, the use of other viruses and viral vectors derived therefrom, including, but not limited to, lentivirus vectors, parvovirus, vaccinia virus, coronavirus, calicivirus, papilloma virus, flavivirus, orthomixovirus, togavirus, picornavirus, adenoviral vectors, modified and attenuated herpesviruses. Any such virus can further be modified with specific surface expressed molecules that target these to HSCs or other stem cells, such as membrane bound SCF, or other stem-cell specific growth factor ligands. Other methods of transfection of mammalian cells include, but are not limited to, direct electroporation of mammalian expression vectors, such as by using NUCLEOFECTOR.TM. technology (AMAXA Biosystems). This technology is a highly efficient non-viral gene transfer method for most primary cells and for hard-to-transfect cell lines, which is an improvement on the long-known method of electroporation, based on the use of cell-type specific combinations of electrical current and solutions to transfer polyanionic macromolecules directly into the nucleus. Additionally, suitable methods of transfection can include any bacterial, yeast or other artificial methods of gene delivery that are known in the art.

[0114] The step of expanding the transfected stem cells or culturing the stem cells and exogenous fusion proteins (e.g., the Tat-fusion proteins described in the variations of this method described below) in the presence of suitable growth factors can include the use of any suitable culture conditions, including those specifically described herein. The combination of suitable stem cell growth factors can include any stem cell factors that allow transfected (e.g., transduced) cells of the invention to grow, survive and proliferate in culture. While specific combinations are described herein, and while this is an important step of the present method, this step can be simply described as providing any combination of growth factors that are suitable for the growth, proliferation and survival of stem cells, and include any combinations that are known in the art. Accordingly, the invention is not limited to a particular combination. One preferred combination of growth factors includes: interleukin-6 (IL-6), IL-3 and stem cell factor (SCF). Another preferred combination of growth factors includes stem cell factor (SCF), thrombopoietin (TPO), insulin-like Growth Factor 2 (IGF-2) and fibroblast Growth Factor 1 (FGF-1), in serum-free media. This latter combination was recently described in Zhang and Lodich (2005; Murine hematopoietic stem cells change their surface phenotype during ex vivo expansion, Blood 105, 4314-20). The stem cells transfected with nucleic acid molecules encoding the combinations proteins described herein (e.g., MYC-ER and Bcl-2 as described in the examples) are expected to also become conditionally immortalized in this cocktail of growth factors, as with the cocktail described in the Examples above (using IL-3, IL-6 and SCF). Other growth factors for use in the invention include, but are not limited to, angiopoietin-like proteins (e.g., Agptl2, Angptl3, Angptl5, Angptl7, etc.), proliferin-2 (PLF2), glycogen synthase kinase-3 inhibitors, inducers of the wnt and Notch signaling pathways, FH3L and related cytokines, fibroblast growth factor 2 (FGF2) and related cytokines, wnt-1 and other activators of the Wnt pathway, Sonic hedghog (shh-1) and other activators of that pathway. Other suitable combinations of growth factors will be applicable to the method of the present invention and will be apparent to those of skill in the art. Indeed, the cell lines generated using the method of the present invention can readily be used to screen for additional cytokines and growth factors that could be used for expanding long-term stem cells, or any of their derived progenitors, in vitro under neutral or directed conditions.

[0115] According to the present invention, a medium suitable for culture of animal cells can include any available medium which has been developed for culture of animal cells and particularly, mammalian cells, or which can be prepared in the laboratory with the appropriate components necessary for animal cell growth, such as assimilable carbon, nitrogen and micronutrients. Such a medium comprises a base medium, which is any base medium suitable for animal cell growth, including, but not limited to, Iscove's Modified Dulbecco's Medium (IMDM), Dulbecco's modified Eagles medium (DMEM), alpha MEM (Gibco), RPMI 1640, or any other suitable commercially available media. To the base medium, assimilable sources of carbon, nitrogen and micro-nutrients are added including, but not limited to, a serum source, growth factors, amino acids, antibiotics, vitamins, reducing agents, and/or sugar sources. It is noted that completed mediums comprising a base medium and many of the additional components necessary for animal cell growth are commercially available, and some media are available for particular types of cell culture. In addition, many serum-free media are available and may be particularly suited for the culture of stem cells according to the invention.

Cells and Compositions

[0116] Another embodiment of the present invention relates to a cell, cell line, or population of cells produced according to the method of the present invention as described herein. Also included in the invention are compositions comprising such cells, cell lines or populations of cells. For therapeutic methods, such compositions can include a pharmaceutically acceptable carrier, which includes pharmaceutically acceptable excipients and/or delivery vehicles, for delivering the cells, cell lines, or cell populations to a patient. As used herein, a pharmaceutically acceptable carrier refers to any substance suitable for delivering a therapeutic composition useful in the method of the present invention to a suitable in vivo site.

Adaptation of the Method of the Invention to Produce Cell Lineages at Intermediate Stages of Development

[0117] Another embodiment of the present invention relates to adaptations of the novel methods described herein to generate cell lines that capture intermediate stages of development for the hematopoietic lineages. According to the present invention, an "intermediate" stage of development or differentiation refers to a pluripotent stage of cell development or differentiation that is downstream of the stage of development or differentiation of the stem cell from which the "intermediate" cell was derived, but is upstream of the final, or terminal, point of differentiation of a cell. For example, a pre-B cell is an intermediate stage of a hematopoietic stem cell, which can still differentiate into a mature B cell. Intermediate stages of development or differentiation will be understood by those of skill in the art.

[0118] More particularly, for many therapeutic and discovery or research applications, as well as for storage of cells lines, it is desirable that the cell lines have a stable phenotype and retain their ability to further differentiate along their committed pathway once the active oncogene with which the cell has been transfected is turned off. Accordingly, the present invention encompasses additional steps of producing cells that have not fully differentiated (are not terminally differentiated), but rather, are at an intermediate stage of differentiation. In one non-limiting example of this embodiment, long-term stem cells produced using the method described above are randomly differentiated in vitro following withdrawal of the conditions that maintain the activity of the protooncogene or other gene that promotes cell survival and proliferation (e.g., 4-OHT in the case of the tamoxifen-dependent protooncogenes), or by applying the appropriate conditions that turn off (inactivate) the protooncogene/oncogene. This step can be performed while maintaining the culture in neutral cytokine growth conditions (e.g., IL-3, IL-6 and SCF), or by replacing those cytokines which could specifically direct differentiation towards a certain lineage (e.g., IL-7 and Notch ligands for lymphoid lineages, GM-CSF and IL-4 for dendritic cells, G-CSF for myelomonocytic cells, etc.) with cytokines that are neutral for differentiation (do not direct or drive differentiation of the cells). Once the cultures begin to display differentiation markers consistent with a specific lineage, the culture media is again supplemented with the conditions that activate the protooncogene (e.g., 4-OHT) or exposed to the conditions that otherwise reactivating the protooncogene, in order to stabilize the phenotype and generate cell lines having a stable, intermediate differentiation phenotype.

[0119] By way of exemplification of this method, the inventors have generated CD4+, .alpha..beta.+ T cells in vitro from ABM42 cells (lt-HSC produced by the method of the invention; see Examples) by withdrawal of 4-OHT from the media, and re-addition of 4OHT after differentiation. The inventors have also generated dendritic cell lines by incubating ABM46 cells (see Examples) in GM-CSF, IL-4 and FLT3L and then placing the cultures back in the presence of 4-OHT after differentiation.

[0120] Another approach for creating such cell lines involves introducing the ctlt-HSC cells into mice to allow for differentiation, and arresting, or stabilizing the phenotypes in vivo after injections of 4-OHT. This method is described in detail in Example 8. Briefly, and by way of example, lt-HSC generated by the present method are injected into immuno-compromised animals (e.g., immuno-compromised mice). The oncogene in the lt-HSCs is reactivated using injections of the activating agent (e.g., 4-OHT), cells are later collected, and then the cells can be cultured in vitro to differentiate the cells, and then stored or used as desired. This approach, and the other described above, can be used for both murine and human ctlt-HSC cell lines, such as by using either NOD/SCID mice as the recipients, or neonatal Rag-1.sup.-/- mice, which will be given intrahepatic injections.

Application of the Method of the Invention to Embryonic Stem Cells

[0121] Another embodiment of the invention relates to the application of the method of conditionally immortalizing stem cells to embryonic stem (ES) cells. Such methods will be useful for generating cell lines that are more readily derived from ES cells, such as cells of the neuronal linage, including neuronal stem cells.

[0122] In this embodiment, the method of the present invention, comprising the transduction of cells (in this case, ES cells) with a protooncogene and a gene that inhibits apoptosis (e.g., MYC-ER and Bcl-2) can be applied to ES cells to further control the directed differentiation of these cells. In this embodiment, such cells can be used to generate transgenic mice, for example, and in addition, any ES cell and relevant progenitor cell population derived therefrom can be subjected to the activation of the protooncogene by exposure to the activating agent, hence allowing for the generation of novel conditionally transformed stem cell lines (different tissue types), or mature cell lines for the tissue type of interest. In addition, the directed differentiation of transduced ES cells in vitro can also be used to capture intermediate states of differentiation by as described above. The use of ES cells or ES-derived cells in this manner provides a novel platform for drug discovery and target identification in the setting of different diseases.

[0123] For example, neuronal stem cells can be employed in this embodiment of the invention, as well as the directed differentiation of ES cells into the neuronal pathway using the method of the invention. The isolation and transduction of neuronal stem cells from the hippocampus has been previously described for mice. The culture conditions for neurospheres would enable the proliferation of those cells, rendering them susceptible to viral-mediated transduction of the genes of the invention (e.g., MYC-ER and Bcl-2), in order to generate conditionally transformed neuronal stem cell lines. Their differentiation in vitro as well as in vivo following implantation can be monitored by virtue of the virally encoded reporter genes as well as previously defined markers of neuronal differentiation. In addition, the administration of the activating agent (e.g., 4-OHT) to the mice following transplantation of the conditionally transformed neuronal stem cell lines may lead to the development of a neurological malignancy (neuroblastoma, glioblastoma, etc.). Those tumors would provide a novel model for preclinical studies and target identification.

[0124] The directed differentiation of ES cells that had been transduced with, for example, MYC-ER and Bcl-2, can be carried out in the presence of a previously defined growth medium, as well as cytokines. The addition of the activating agent (e.g., 4-OHT) at any time during the culture will enable the stabilization of the cells at an intermediate phenotype, and leads to the generation of cell lines that still retain the capacity to undergo further differentiation. For instance, the generation of dopaminergic neurons from ES cells is normally done by the addition of Retinoic acid and FGF8. This type of neuron would be ideal for repairing brain lesions observed in Alzheimer's patients. However, the transplantation of fully differentiated neuronal cells may preclude their successful implantation and engraftment. A conditionally transformed cell line that was committed to the dopaminergic neuronal pathway, but still retained its ability to further differentiate after transplant, as envisioned herein, is expected to greatly increase the chances of implantation and successful engraftment. A similar scenario can be proposed for the generation of motor neurons from ES cells, by adding Retinoic acid and a sonic hedgehog agonist to the cultures. Those neuronal cells could help repair spinal cord injuries. Once again, fully differentiated cells would not be used in this embodiment, but rather, the committed progenitor cells that retain the capacity to differentiate (produced by the method of the invention) would be employed.

Variations or Modifications of the Method of Conditional Immortalization for the Removal of the Transgene

[0125] In one embodiment of the invention, in order to avoid taking the risk of introducing stem cells that harbor transgenes such as those described herein (e.g., MYC-ER) into humans and/or mice, the recombinant constructs are designed so that these DNA fragments will be excised. This embodiment can be achieved using any suitable method of first establishing the long-term stem cells according to the method of the invention, and then exposing the cells (or a patient) to conditions under which the recombinant DNA will be removed, excised or completely silenced.

[0126] For example, in one aspect of the invention, a bacterial recombinase approach is used. In this aspect of the invention, preferably, two different recombinases are used in order to allow control over which one of the two genes is excised at any one point in time. Two examples of such recombinases are the Cre and Flp recombinases, which are well-known in the art. Briefly, the recognition substrate sequences (RSS's) for one of the recombinases is introduced into the retroviral constructs such that they flank the open reading frame of the oncogene, as well as the reporter gene (e.g., GFP or Thy1.1). In this case, the cells are incubated in media containing a Tat-Cre fusion protein (i.e., HIV or other retroviral Tat protein fused to Cre). This recombinant protein has been previously described and shown to be able to passively enter cells, and mediate loxP site-dependent recombination of genomic DNA. Other methods of gene (nucleic acid molecule) excision are known to those of skill in the art and could readily be applied to the present invention. Examples 5 and 13 exemplify this embodiment of the invention.

[0127] In another embodiment of the invention, to provide another method of avoiding the risk of introducing stem cells that harbor transgenes such as those described herein into humans and/or other animals (e.g., mice), instead of transfecting the stem cells with the combination of the recombinant constructs for the protooncogene or the anti-apoptosis protein, the invention is performed by making use of Tat-fusion proteins as a method to allow the proteins access to the inside of the cell without having to introduce transgenes into the cell. For example, recombinant constructs that encode tat-protooncogene or tat-anti-apoptosis genes (e.g., Tat-MYC-ER or Tat-Bcl-2) may be used to conditionally immortalize stem cells. In this embodiment of the invention, the target stem cells will be cultured under suitable culture conditions, in media that contains purified recombinant Tat-fusion proteins encoded by the specific gene combination selected (e.g., MYC-ER and Bcl-2). In this embodiment of the invention, the protooncogene product or similar gene product can be inducible, as in the embodiments above. Alternatively, or in addition, the action of this protein can be regulated simply by providing or removing the protein from the culture. While the cell lines that are generated with this approach will be continuously dependent upon the addition of the exogenous Tat-fusion proteins, they will not have a specific exogenous nucleotide sequence introduced into them. The absence of foreign oncogene sequences is expected to improve the clinical deployment of the method of the present invention. Human immunodeficiency virus-1 (HIV-1) Tat, is one exemplary Tat protein, although other retroviral Tat proteins are known in the art. As a non-limiting example, the nucleic acid sequence encoding HIV-1 Tat is represented herein as SEQ ID NO:9, which encodes an amino acid sequence represented herein by SEQ ID NO:10.

[0128] In another embodiment, to provide another method of avoiding the risk of introducing stem cells that harbor transgenes such as those described herein into humans and/or other animals (e.g., mice), instead of transfecting the stem cells with the combination of the recombinant constructs for the protooncogene or the anti-apoptosis protein, the invention is performed by introducing proteins (e.g., MYC and Bcl-2) into a cell using aptamer technology. Aptamers are short strands of synthetic nucleic acids (usually RNA but also DNA) selected from randomized combinatorial nucleic acid libraries by virtue of their ability to bind to a predetermined specific target molecule with high affinity and specificity. Aptamers assume a defined three-dimensional structure and are capable of discriminating between compounds with very small differences in structure. Accordingly aptamers can be conjugated with the proteins used in the invention or with non-integrating cDNA encoding the proteins, for example, and used to deliver the proteins or DNA to the cells. In addition, aptamers can readily be used to deliver siRNA to cells, for example, when one disrupts proapoptotic proteins according to the present invention. Aptamer technology is discussed, for example, in Davidson, 2006, Nature Biotechnol. 24(8):951-952; and McNamara et al., 2006, Nature Biotechnol. 24(8):1005-1015). Again, the absence of foreign oncogene sequences is expected to improve the clinical deployment of the method of the present invention.

[0129] In another embodiment, to provide another method of avoiding the risk of introducing stem cells that harbor transgenes such as those described herein into humans and/or other animals (e.g., mice), instead of transfecting the stem cells with the combination of the recombinant constructs for the protooncogene or the anti-apoptosis protein, the invention is performed by introducing the protooncogene and/or anti-apoptosis protein into a cell using CHARIOT.TM. technology (Krackeler Scientific, Inc., Albany, N.Y.). With this technology, a non-covalent bond is formed between a CHARIOT.TM. peptide and the protein of interest. This protects the protein from degradation and preserves its natural characteristics during the transfection process. Upon delivery to a cell, the complex dissociates and CHARIOT.TM. is transported to the nucleus, while the delivered protein is biologically active and free to proceed to its cellular target. Efficient delivery can occur in the presence or absence of serum, and is independent of the endosomal pathway, which can modify macromolecules during internalization. This delivery system also bypasses the transcription-translation process. Accordingly, the proteins useful in the present invention can be delivered to a cell and released to conditionally immortalize the cell, without the need for the introduction of a protooncogene or oncogenes to the cell. As above, the absence of foreign oncogene sequences is expected to improve the clinical deployment of the method of the present invention.

[0130] As yet another alternative (or additional) means to control for the possibility of an insertion of a protooncogene into the host cell genome by the various viral approaches described herein, and thereby avoid a transforming event, a drug sensitivity (drug susceptibility) cassette can be introduced into the viral constructs to be used such that it will be expressed in every transduced cell and its differentiated progeny. A drug sensitivity cassette or a drug susceptibility cassette is a nucleic acid sequence encoding a protein that renders a cell susceptible or sensitive to the presence of a particular drug, so that upon exposure to the drug, the cell activity is inhibited and preferably, undergoes apoptosis. Those patients in which the levels of a particular blood cell population increases without apparent cause (e.g., infection, trauma, stress, etc.), can be given a course of the drug to which sensitivity has been introduced in order to ablate those cells and mitigate any possible additional complications involving cells in which the genetic insertions may have inadvertently caused an oncogenic mutation. Accordingly, as a non-limiting example, one could introduce into a construct used in the method of the invention a cassette that encodes the cDNA for HPRT in order to render the transduced cells susceptible to 6-thioguanine. Another non-limiting example is the introduction of the thymidine kinase cDNA from a Herpes-simplex virus family member (HSV-TK), in order to render the transduced cells susceptible to relevant inhibitors such as Ganciclovir, Acyclovir, and any relevant derivatives. In addition, any other such drug sensitivity cassettes and their relevant agonists would work in this context.

[0131] Other methods of introducing nucleic acids or proteins according to the present invention into a cell will be apparent to those of skill in the art. Those that minimize or eliminate the risk of introducing recombinant DNA into a host cell genome are preferred by the invention, many such examples being described above.

Methods of Use for Conditionally Immortalized Cells of the Invention

[0132] Another embodiment of the present invention includes any of the stem cell populations, including mixed and clonal populations, that are produced by the method of the invention, as well as the use of the stem cells of the invention in any of the methods described herein, including differentiation into a desired cell type, and any method of transplantation, cell replacement, disease therapy, genetic engineering, drug discovery, and investigation of cell development and differentiation as described herein.

[0133] Since one can now produce virtually unlimited supplies of homogeneous stem cells that can readily be stored, recovered, expanded and manipulated, such stem cells can be used as stem cells or differentiated into various cell lineages and used in assays to test various compounds for effects on cell differentiation, gene expression, and cell processes. Therefore, one embodiment of the invention relates to a method to identify compounds that effect cell differentiation, gene expression, and/or cell processes. The method generally includes the steps of contacting stem cells produced by the method of the present invention with a compound to be tested, and measuring a particular result, and particularly a desired result, such as gene expression, a biological activity, cell differentiation, cell growth, cell proliferation, etc. (see below), as compared to in the absence of the compound, to determine whether or not the test compound had the desired effect on the stem cell. This method can be used to test for virtually any aspect of cell differentiation, cell activity or gene expression. In one aspect, the stem cells are manipulated prior to contact with the compounds, such as by genetic manipulation. Stem cells from individuals with genetic defects can be evaluated in such assays in order to identify therapeutic compounds (e.g., cancer therapeutics) and to evaluate gene replacement therapies, for example. Indeed, the technology of the present invention provides an opportunity to target the cells of a specific individual to identify drug candidates and therapeutic candidates and strategies that are "tailored" to the cells of an individual. Furthermore, as discussed above, such assays can also be used to identify other growth factors or culture conditions that are suitable for maintaining the stem cells of the invention in culture. An example of such an assay is described in detail below in Example 7, although the invention is not limited to this assay.

[0134] Another embodiment of the invention relates to a method to study cell lineage commitment and/or differentiation and development of cells from a stem cell, which generally comprises culturing the conditionally immortalized stem cells of the present invention and evaluating such cells for genetic and biological markers related to cell development and differentiation under various conditions and in the presence and absence of compounds or agents that may affect cell lineage commitment or differentiation. As discussed above, prior to the present invention, such studies were severely hampered by the lack of access to and the inability to generate sufficient numbers of the desired cell population to perform desired experiments. For example, in order to identify or screen for intermediates in the differentiation of a particular progenitor cell line, a sufficient number of cells must be obtained to provide meaningful and reproducible results. Using technologies available at the time of the invention, this was not possible. However, the present invention solves the problem by providing expandable and essentially unlimited supplies of homogeneous stem cells that can be used in a variety of experiments. This technology will greatly enhance research capabilities in the area of cell differentiation and discovery. In one aspect, conditionally immortalized stem cells of the invention are expanded, and then a subset are cultured in the absence of the conditions that maintain the cells in the conditionally immortalized state (e.g., in the absence of tamoxifen, according to the exemplary method illustrated herein). The cells can be evaluated for changes in gene expression, cell surface markers, secretion of biomolecules, or any other genotypic or phenotypic marker, to study the process of cell differentiation and lineage commitment. Growth factors or other factors can be added to the cultures, for example to drive differentiation down a particular cell lineage pathway, and the changes in the cells can be evaluated in the presence or absence of such factors. Furthermore, the cells can be used to evaluate culture conditions, in vivo conditions, factors, and agents that influence (regulate) cell differentiation and development.

[0135] Various methods of detection of changes in genotypic or phenotypic characteristics of cells in any of the assays of the invention are known in the art. Examples of methods that can be used to measure or detect gene sequence or expression include, but are not limited to, polymerase chain reaction (PCR), reverse transcriptase-PCR (RT-PCR), in situ PCR, quantitative PCR (q-PCR), in situ hybridization, Southern blot, Northern blot, sequence analysis, microarray analysis, detection of a reporter gene, or other DNA/RNA hybridization platforms. Methods to measure protein levels, include, but are not limited to: Western blot, immunoblot, enzyme-linked immunosorbant assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, surface plasmon resonance, chemiluminescence, fluorescent polarization, phosphorescence, immunohistochemical analysis, matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry, microcytometry, microarray, microscopy, fluorescence activated cell sorting (FACS), flow cytometry, and assays based on a property of the protein including but not limited to DNA binding, ligand binding, interaction with other protein partners, cell signal transduction, enzyme activity, and secretion of soluble factors or proteins.

[0136] In drug screening assays, the term "test compound", "putative inhibitory compound" or "putative regulatory compound" refers to compounds having an unknown or previously unappreciated regulatory activity in a particular process. As such, the term "identify" with regard to methods to identify compounds is intended to include all compounds, the usefulness of which as a compound for a particular purpose (e.g., regulation of cell differentiation) is determined by a method of the present invention, preferably in the presence and absence of such a compound. Compounds to be screened in the methods of the invention include known organic compounds such as antibodies, products of peptide libraries, and products of chemical combinatorial libraries. Compounds may also be identified using rational drug design. Such methods are known to those of skill in the art and can involve the use of three-dimensional imaging software programs. For example, various methods of drug design, useful to design or select mimetics or other therapeutic compounds useful in the present invention are disclosed in Maulik et al., 1997, Molecular Biotechnology: Therapeutic Applications and Strategies, Wiley-Liss, Inc., which is incorporated herein by reference in its entirety.

[0137] In any of the above-described assays, the conditions under which a cell, cell lysate, nucleic acid molecule or protein of the present invention is exposed to or contacted with a putative regulatory compound, such as by mixing, are any suitable culture or assay conditions, which can include the use of an effective medium in which the cell can be cultured (e.g., as described above) or in which the cell lysate can be evaluated in the presence and absence of a putative regulatory compound. Cells of the present invention can be cultured in a variety of containers including, but not limited to, tissue culture flasks, test tubes, microtiter dishes, and petri plates. Culturing is carried out at a temperature, pH and carbon dioxide content appropriate for the cell. Such culturing conditions are also within the skill in the art, and particularly suitable conditions for culturing conditionally immortalized stem cells of the present invention are described in detail elsewhere herein. Cells are contacted with a putative regulatory compound under conditions which take into account the number of cells per container contacted, the concentration of putative regulatory compound(s) administered to a cell, the incubation time of the putative regulatory compound with the cell, and the concentration of compound administered to a cell. Determination of effective protocols can be accomplished by those skilled in the art based on variables such as the size of the container, the volume of liquid in the container, conditions known to be suitable for the culture of the particular cell type used in the assay, and the chemical composition of the putative regulatory compound (i.e., size, charge etc.) being tested.

[0138] In one embodiment of the invention, the cells and methods of the invention are useful for methods directed at evaluating pluripotency of ctlt-HSCs derived from human cord blood, CD34+ cells, or adult CD34+ cells isolated from peripheral blood. Such a method is described in Example 11.

[0139] Yet another embodiment of the invention relates to the use of ctlt-HSC cell lines as a platform to generate novel models of Acute Myeloid Leukemia (AML). More particularly, the present inventors have generated a mouse model of acute myeloid leukemia using the ctlt-HSCs of the invention. These are leukemias composed of cells that resemble HSCs, based on their surface marker expression. In order to generate ctlt-HSCs to promote leukemia in mice, 10.sup.3-10.sup.5 ctlt-HSCs are transferred along with 10.sup.5 Rag-1.sup.-/- whole bone marrow cells into lethally irradiated recipient mice. The mice are given weekly doses of 4-OHT in order to maintain oncogene activity, and monitored for clinical signs associated with leukemia, as known in the art. Tumors have been recovered from these animals and they can be propagated in culture in the absence of 4-OHT. Those cells retain their HSC-like phenotype, indicating that they are no longer exquisitely dependent upon MYC hyperactivity in order for proliferation, survival and arrested differentiation. The leukemic cell lines can also confer the disease upon secondary transplantation to irradiated recipient mice. These tools provide a novel platform for studying the biology and exporting new therapeutic avenues for AML and related diseases. Furthermore, the introduction of ctlt-HSC cell lines into mice that are treated with 4-OHT will provide a good built-in positive control for therapy: the withdrawal of 4-OHT. The secondary cell lines that arose after the establishment of tumors in vivo can also be used to understand the relevant therapeutic targets for drug resistant forms of AML.

[0140] Other embodiments of the present invention relate to the use of the stem cells generated by the method of the present invention, as well as cells differentiated from those stem cells, in a variety of therapeutic and health-related methods. These methods generally include the steps of obtaining a population, culture or line of conditionally immortalized stem cells produced by the method of the present invention, removing the conditions under which such cells are conditionally immortalized, and then using the cells in a therapeutic protocol. For example, the cells can be administered directly to an individual in need of the cells or the cells can be differentiated into a desired cell type in vitro and then administered to an individual. In addition, prior to or just after the removal of the conditions under which the cells are immortalized, the cells can be genetically modified in vitro to express or silence a gene or genes, as a novel method of gene therapy under a controlled environment. The cells can then be administered to an individual as stem cells or first differentiated in vitro to a desired cell lineage.

[0141] To obtain the stem cells, in one embodiment, stem cells are obtained from the individual to be treated, and are then conditionally immortalized according to the method of the invention. These cells can be expanded extensively, stored (e.g., frozen or cryopreserved), and then retrieved and expanded again, manipulated, and/or used repeatedly as required. In another embodiment, one obtains the stem cells by accessing a previously stored source of conditionally immortalized stem cells from the individual to be treated. In yet another embodiment, the stem cells are obtained from a panel of human stem cell lines that were previously generated and which cover a significant percentage of the population according to the current criteria used to identify "matching" donors. In one embodiment, the cells are obtained from fresh, or cryopreserved cord blood, hematopoietic progenitor populations that can be derived from the directed differentiation of ES cells in vitro, HSCs obtained from the peripheral blood of normal, or G-CSF treated patients who have been induced to mobilize their lt-HSCs to the peripheral circulation. Other sources of stem cells will be apparent to those of skill in the art. The cells are cultured according to the methods described previously herein and the conditions controlling immortalization can be removed at the appropriate time. In addition, prior to administration of the cells to an individual, the cells can be manipulated to excise the genes or constructs that are responsible for the conditional immortalization (i.e., the protooncogene and/or the anti-apoptosis encoding gene), or if the cells are maintained through the use of soluble fusion proteins in the culture medium, as described above for the Tat-fusions, these soluble proteins can be removed from the culture gradually or immediately.

[0142] Therefore, the present invention includes the delivery of stem cells produced by the method of the invention (including compositions comprising such stem cells), or cells differentiated from these cells, to an individual (which can include any animal). Since the stem cells used in these methods are produced in vitro, even if stem cells were initially isolated from the patient, the entire administration process of the cells is essentially an ex vivo administration protocol. Ex vivo administration refers to performing part of the regulatory step outside of the patient, such producing the conditionally immortalized stem cells that were removed from an individual (which can include producing genetically modified stem cells in addition to essentially normal stem cells), and returning the cells, or cells differentiated from these cells, to the patient. The stem cells produced according to the present invention or cells differentiated therefrom can be returned to an individual, or administered to an individual, by any suitable mode of administration. Such administration can be systemic, mucosal and/or proximal to the location of a target site. The preferred routes of administration will be apparent to those of skill in the art, depending on the type of condition to be prevented or treated or the reason for administration. Preferred methods of administration include, but are not limited to, intravenous administration, intraperitoneal administration, intramuscular administration, intranodal administration, intracoronary administration, intraarterial administration (e.g., into a carotid artery), subcutaneous administration, transdermal delivery, intratracheal administration, subcutaneous administration, intraarticular administration, intraventricular administration, intraspinal, pulmonary administration, impregnation of a catheter, and direct injection into a tissue (e.g., such as cannulation of the liver, for example).

[0143] The cells can be administered with carriers or pharmaceutically acceptable excipients. Carriers are typically compounds that increase the half-life of a therapeutic composition in the treated individual. Suitable carriers include, but are not limited to, polymeric controlled release formulations, biodegradable implants, liposomes, oils, esters, and glycols. As used herein, a pharmaceutically acceptable excipient refers to any substance suitable for delivering cells produced by the method of the present invention to a suitable in vivo site. Preferred pharmaceutically acceptable excipients are capable of maintaining a cells in a form that, upon arrival of the cells at a target tissue or site in the body, the cells are capable of functioning in a manner that is beneficial to the individual.

[0144] According to the present invention, an effective administration protocol comprises suitable dose parameters and modes of administration that result in delivery of a useful number of functional cells to a patient in order to provide a transient or long-term benefit to the patient. Effective dose parameters can be determined using methods standard in the art for a particular condition or disease. Such methods include, for example, determination of survival rates, side effects (i.e., toxicity) and progression or regression of disease.

[0145] A suitable single dose of stem cells or cells differentiated therefrom according to the present invention is a dose that is capable of providing a beneficial number of cells to a patient, when administered one or more times over a suitable time period. For example, a preferred single dose of stem cells according to the present invention is from about 0.5.times.10.sup.4 to about 5.5.times.10.sup.8, or from about 0.5.times.10.sup.5 to about 5.5.times.10.sup.7, or from about 0.5.times.10.sup.6 to about 5.5.times.10.sup.10 stem cells per individual per administration, with doses from about 1.times.10.sup.8 to about 5.5.times.10.sup.10 being even more preferred. Any dose in between 0.5.times.10.sup.4 and about 5.5.times.10.sup.10 is encompassed by the invention, in increments of 10.sup.2 cells. Higher or lower doses will be known to those of skill in the art depending on the type of stem cell or differentiated cell to be administered, and also depending on the route of administration. It will be obvious to one of skill in the art that the number of doses administered to an animal is dependent upon the extent of the condition or disease and the response of an individual patient to the treatment. Thus, it is within the scope of the present invention that a suitable number of doses includes any number required to treat a given disease.

[0146] As used herein, the phrase "protected from a disease" refers to reducing the symptoms of the disease; reducing the occurrence of the disease, and/or reducing the severity of the disease. Protecting an animal (an individual, a subject) can refer to the ability of cells produced according to the present invention, when administered to an animal, to prevent a disease from occurring and/or to cure or to alleviate disease symptoms, signs or causes. As such, to protect an animal from a disease includes both preventing disease occurrence (prophylactic treatment) and treating an animal that has a disease or that is experiencing initial symptoms of a disease (therapeutic treatment). The term, "disease" refers to any deviation from the normal health of a mammal and includes a state when disease symptoms are present, as well as conditions in which a deviation (e.g., infection, gene mutation, genetic defect, etc.) has occurred, but symptoms are not yet manifested.

[0147] As discussed above, the stem cells of the present invention can be administered to an individual to treat or prevent a variety of conditions. For example, the stem cell lines of the present invention provide a unique source of expandable stem cells for use in a variety of transplantation and therapeutic strategies, including the treatment of cancer, and particularly, cancer that is treated by radiation. In addition, a variety of immune deficiency disorders and anemia disorders (e.g., aplastic anemia or hemolytic anemia) will also benefit greatly from this technology, since the present invention provides the ability to repopulate hematopoietic cells of an individual as needed by the individual. Another application of the present invention relates to the generation of continuously expandable and renewable hair follicle stem cells, for use, for example in the context of reconstructive surgery for burn victims, for any individual that undergoes chemotherapy and/or radiation therapy resulting in the irreversible loss of hair growth, as well as patients following any surgical procedure affecting the skull or in elective procedures that involve the induction of hair growth in individuals affected by hereditary pattern baldness. Similarly, application of the present invention to stem cells of the skin will be invaluable for use in wound healing and treatment of burn victims, as well as plastic reconstructive surgery for trauma and other patients, as well as elective surgeries, including, but not limited to, cosmetic surgery. Such cells can be additionally genetically manipulated to correct inborn or acquired genetic defects in young and aged individuals. One of skill in the art will understand based on this disclosure that benefits can be derived from the use of the present invention on various other stem cell populations, including, but not limited to, stem cells derived from lung, breast, and intestinal epithelium and stem cells derived from neural and cardiac tissue, to name just a few.

[0148] In addition, as discussed above, the present invention provides the unique opportunity for an individual to have access to expandable supplies of autologous stem cells and cells differentiated therefrom as needed throughout the life of the individual. Such stem cells generated by the present method can be stored and used as part of therapeutic protocols during the lifetime of the individual, should they be needed (e.g., in the event the individual develops a cancer or immune deficiency disease).

[0149] Genetic defects can now be corrected or beneficial gene modifications can be introduced into somatic cells by manipulating autologous stem cells obtained from an individual that have been conditionally immortalized and expanded using the method of the present invention. The stem cells can then be reintroduced into the individual from whom they were obtained.

[0150] Additional applications of the present invention include the use of stem cell lines to repair lung injury that occurs as a result of COPD, IPF, emphysema, asthma and smoking. In addition, such cells could be used to treat blood vessel damage in the heart, and help in autoimmune diseases after lethal irradiation (e.g., SLE, diabetes, RA).

[0151] In the method of the present invention, cells produced according to the method of the invention and compositions comprising the cells can be administered to any animal, including any member of the Vertebrate class, Mammalia, including, without limitation, primates, rodents, livestock and domestic pets. A preferred mammal to treat is a human.

[0152] Various aspects of the present invention are described in more detail in the following Examples and the attached figures. However, the present invention is not limited to these examples and illustrations of the invention.

EXAMPLES

Example 1

[0153] The following example describes the development of a method to reversibly immortalize long-term hematopoietic stem cells (lt-HSCs).

[0154] Elucidation of the molecular basis of the impairment in hematopoietic lineage development has been complicated historically by the low frequency of relevant cell populations, which prevents biochemical analysis of signaling and downstream responses. In fact, this has been a major limiting factor in all studies of hematopoiesis. In addition, the limited availability of LT-HSCs has also been a major obstacle in the treatment of many types of cancer as well as several kinds of immune deficiencies in humans.

[0155] In an effort to overcome this limitation, the present inventors developed a method to produce conditionally transformed cell lines representing early hematopoietic stem cell progenitors. The initial strategy involved retroviral transduction of bone marrow stem cells from 5FU treated young and immunologically aged 3-83 mice. The inventors utilized the pMSCV bisistronic retroviral vector with inserts encoding Bcl-2 and GFP, and MYC-ER and GFP [Van Parijs, L., Y. Refaeli, A. K. Abbas, and D. Baltimore. (1999) Autoimmunity as a consequence of retrovirus-mediated expression of C-FLIP in lymphocytes. Immunity, 11, 763-70]. These genes were selected because the present inventors knew that MYC has the ability to replace cytokine derived survival and proliferative signals in lymphocytes. By restricting the target cell, the inventors hypothesized that stem cell tumors might form. Importantly, MYC-ER function is tamoxifen dependent in this setting, allowing the termination of MYC function and transformation by withdrawing tamoxifen from the animal or cultures. In cells transduced with MYC-ER, the fusion protein is produced, but is retained in the cytoplasm until exposed to tamoxifen.

[0156] More specifically, stem cell populations from 5FU treated mice were transduced with both retroviruses (encoding MYC-ER and Bcl-2) and transferred into lethally irradiated recipient mice (1200 rads). Ten days later, weekly intraperitoneal injections of 1 mg/mouse of 4-hydroxytamoxifen (40HT) emulsified in oil were initiated to activate MYC function (FIG. 1). Within four weeks, recipients of young (but not old) transduced stem cells developed tumors. The tumors were harvested from bone marrow, spleen and lymph nodes and cultured in vitro with tamoxifen, but without added cytokines. These cells grew for about 10 days, but then growth stopped and the cells eventually died. the inventors suspected that the cells were differentiating and considered that this might have been due to requirements for cytokines for growth of the cells. Referring to FIG. 1, the curves represent the kinetics of mortality after transplantation and activation of MYC function in vivo. The mice uniformly succumbed to leukemias. While the overexpression of MYC can replace the cytokine-dependent proliferation and survival function, it does not seem to be involved in the cytokine-derived differentiation signals.

[0157] When ill, the mice were euthanized. Bone marrow, spleen and lymph node cells were harvested and placed in culture with tamoxifen and a stem cell growth factor cocktail (IL-6, IL-3 and stem cell factor (SCF)). In parallel, cells were analyzed by flow cytometry (FIG. 2). Referring to FIG. 2, the dot plots represent the flow cytometric data for the forward (FSC) and side (SSC) scatter characteristics of the HSCs after three days in culture with IL-3, IL-6 and SCF. These two criteria correlate with cell size (FSC) and granularity (SSC). The two populations have similar profiles. The histograms represent the levels of GFP expressed in each cell population. This reflects the efficiency of retroviral transduction in vitro with retroviruses that encode cDNAs for MYC-ER and Bcl-2.

[0158] In all cases, ex vivo GFP.sup.+ cells were >90% Sca-1.sup.+ and Lineage marker negative. After a few days in culture, cells began to grow and approximately 400 lines were frozen for later study. After propagation, these cells retained expression of EGFP and were homogeneously positive for SCA1 and negative for CD34, F1k2 and lineage markers (FIG. 3). The only difference in marker expression between young mouse-derived and aged mouse-derived markers was increased expression of c-kit in young. Without being bound by theory, the present inventors believe that this may have resulted from longer culture (3 months vs. 3 weeks) of aged lines in c-kit ligand before markers were analyzed. Finally, the inventors discovered that these lines can be recovered easily after freezing and retained their original phenotype. Importantly, these cell lines are homogenous in phenotype and exhibit the phenotype of lt-HSC that provide all long term reconstitution in mice (Reya, T., Duncan, A. W., Ailles, L., Domen, J., Scherer, D. C., Willert, K., Hintz, L., Nusse, R., and Weissman, I. L. (2003). A role for Wnt signaling in self-renewal of hematopoietic stem cells. Nature 423, 409-14).

[0159] Recently, the inventors thawed 10 bone marrow derived lines produced as described above, and were able to recover 9 out of 10 of these lines easily by culture in the cytokine cocktail and 4OHT. The inventors phenotyped these tumors, and the results were extremely promising. Specifically, each line contained two distinct cell populations based on forward and 90.degree. light scatter. The nine lines differed only in the proportionality of these populations. The larger of these populations in cell size were uniformly GFP bright and positive for Sca1, Endoglin and ckit but negative for Flt3, B220, CD19 and mIgM. CD34.sup.- also appeared to be negative, although this required confirmation (FIGS. 3A and 3B). This phenotype corresponds perfectly with the published characteristics of long term repopulating pluripotent stem cells (Reya et al., supra). The inventors observed the same initial phenotype on the cell lines that they recently obtained from leukemias that developed from transduced HSCs obtained from young donor mice (FIGS. 3A and 3B).

[0160] To test the ability of these cells to differentiate, representative lines were cultured with and without tamoxifen and in the presence of IL-3, IL-6 and SCF to terminate MYC-ER function for 7 days before analyzing phenotypic markers. As shown in FIG. 4, a significant proportion of cells acquired B lineage markers including B220 (.about.12%), CD19 (.about.10%) and mIgM (.about.10%). In addition, the inventors have been able to generate the following lineages in vitro by withdrawal of 4OHT from the cultures: CD4+ab T-cells, myeloid cells (Mac-1+), ter-119+ erythroid progenitor cells, NK1.1 expressing cells, neutrophils (Gr-1+ cells). Further experiments will assess the ability of these cells to give rise to other lineages, as well as the effect of altering the cytokine regimen on differentiation. Although the comparison has not been performed, the present inventors expect differentiation from young animals, as compared to aged animals to be much more efficient in B cell production. To the best of the present inventors' knowledge, this is the first example of a conditionally immortal hematopoietic stem cell line that can be induced to differentiate in vitro.

Example 2

[0161] The following example describes the results of adoptive transfer of LT-HSC lines into lethally irradiated recipients.

[0162] If the HSC lines described in Example 1 are to be appropriate subjects for analysis of the basis of defective B cell lymphopoiesis in aged animals, they should recapitulate the defect in vivo. The inventors have begun to address this question by adoptive transfer of LT-HSC lines into lethally irradiated recipients. In initial experiments, lines from aged animals (>60% ID.sup.-) were transferred along with RAG2.sup.-/- bone marrow, and recipients were not treated with tamoxifen in order to silence MYC-ER. Six weeks later recipient bone marrow and spleen cells were harvested and the recovery and phenotype of GFP.sup.+ cells (GFP marks cells derived from HSC lines) was analyzed (FIGS. 5A and 5B).

[0163] In the data from three mice presented in FIGS. 5A and 5B, one mouse received the aged HSC line ABM42, and two mice received aged HSC line ABM46. Depending upon the line transferred, 30 to 70% of cells in the lymphoid scatter gate were GFP.sup.+. As shown in FIG. 5, both lines tested (ABM46 and ABM42) gave rise to B (CD19.sup.+) and T (TCR.sup.+, CD4.sup.+, CD8.sup.+) cells, macrophages (CD11b.sup.+) and granulocytes (GR1.sup.+). There was some recipient to recipient variation in the proportionality of these lineages. However, importantly, while both lines tested gave rise to mature CD4 and CD8 single positive T cells (FIG. 7), B cell development did not proceed beyond the progenitor stage (FIG. 6). While B220.sup.+, CD19.sup.+ cells developed, they did not progress to the mIg.sup.+ stage. This is precisely the outcome predicted by results of experiments involving autoreconstitution and adoptive reconstitution using BM HSC from immunologically aged mice (Johnson, S. A., S. J. Rozzo, and J. C. Cambier, Aging-dependent exclusion of antigen-inexperienced cells from the peripheral B cell repertoire. J Immunol, 2002. 168(10): p. 5014-23). In other words, the same developmental arrest is observed when whole bone marrow from immunologically aged mice is used for transplantation.

[0164] The inventors have found that this system can be taken a step further, successfully re-establishing LT-HSC lines from bone marrow of adoptive recipients of the original HSC lines (data not shown). This was accomplished simply by culturing bone marrow cells in stem cell cytokines plus tamoxifen to reactivate MYC. These cells are now growing and exhibit the original phenotype.

Example 3

[0165] The following example describes a method for reversibly immortalizing HSCs using a method conducted entirely in vitro.

[0166] In addition to the method for generating conditionally immortalized long term HSC cell lines described previously herein, the inventors have been able to carry out this procedure completely in vitro. The method described above relies upon introducing the transduced HSC's into mice, and inducing their transformation in vivo. The advantage of carrying this procedure out in vitro is that every aspect of the process is carried out in a controlled environment.

[0167] The method first includes the treatment of donor mice with 5-fluorouracil (5-FU) in order to enrich for HSCs and induce these cells to proliferate. 5FU enriched hematopoietic stem cells from the tibia and femurs of mice were collected and then plated in 24 well tissue culture plates in DMEM media containing 15% heat inactivated fetal calf serum and IL-3, IL-6 and SCF, at a density of 1.8-2.0.times.10.sup.6 cells per well. The cells were subjected to three rounds of spin infection in order to retrovirally transduce the cells with retroviral vectors encoding MYC-ER and Bcl-2. Briefly, the cells were transfected with pMIG-MYC.ER or pMIT-Bcl2. The virus containing supernatants were collected and supplemented with 4 .mu.g/ml of polybrene and 10 mM HEPES, and passed through a 0.45 .mu.m filter. The two different viral supernatants were mixed at a 1:1 ratio and added to the wells. The cells were then centrifuged at 2000 rpm for one hour. The viral supernatants were replaced at the end of each spin infection. 24 hours after the last round infection, the levels of transduction were determined by flow cytometric analysis in order to determine the transduction efficiency. The transduced cells were then incubated in DMEM medium containing IL-3, IL-6, SCF and 10 nM 4OHT. The medium was replaced every 3 days and special emphasis was placed on ensuring a fresh supply of cytokines and 4OHT. Cells are passed slowly, and as needed.

[0168] Using this in vitro approach, the inventors have been able to generate conditionally immortalized cell lines with the following combinations of genes: MYC-ER and Bcl-2; MYC-ER and hTERT (reverse transcriptase component of the human telomerase); ICN-1-ER (ER-regulated active element of the intracellular portion of Notch-1) and Bcl-2; ICN-1-ER and hTERT; and MYC-ER and ICN-1-ER. The data presented in FIGS. 8-11 show the initial characterization of most of these cell lines. They yielded lines composed of c-kit+, Sca-1+, CD34-, flk2- cells, which is a phenotype that is consistent with the one presented by normal long-term hematopoietic stem cells. The data presented in FIGS. 8-11 is derived from the flow cytometric analysis of retrovirally encoded reporter genes (GFP and thy1.1), as well as four markers for stem cells: c-kit, sca-1, CD34 and flk-2. The cell lines shown in FIGS. 8-11 had been in culture for 5 weeks prior to phenotyping. These cells have been expanded and divided in continuous culture for over 35 days to date.

[0169] Referring to FIG. 8, this figure shows the phenotypic comparison of cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with BCL-2 and MYC-ER and maintained in continuous in vitro culture for >90 days. Shown is the phenotype of representative clones 3 (young) months after 90 days of continuous of culture.

[0170] Referring to FIG. 9, this figure shows the phenotypic comparison of cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with different combinations of oncogenes and maintained in continuous in vitro culture for >90 days. 5FU enriched HSCs were retroviral transduced with pMIG-MYC and pMIT-Bcl-2 (top panels), pMIG-MYC.ER and pMIG-hTERT (middle panels), or pMIG-ICN.1.ER and pMIT-Bcl-2. The cells were maintained in DMEM supplemented with 15% fetal calf serum, and a cocktail of IL-6, IL-3 and SCF. Shown is the phenotype of representative clones 3 (young) months after 90 days of continuous of culture. The panels represent the results of the flow cytometric analysis for expression of the viral expression markers (GFP and Thy1.1), as well as four markers required to define long-term HSCs in mice, Sca-1, c-kit, CD34 and Flk-2. The four cell lines contained subpopulations that retained the phenotypes of lt-HSCs (Sca-1+, c-kit+, CD34-, flk-2-).

[0171] Referring to FIG. 10, this figure shows the phenotypic comparison of cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with different combinations of oncogenes and maintained in continuous in vitro culture for >90 days. 5FU enriched HSCs were retroviral transduced with pMIG-ICN.1.ER and pMIT-Bcl-2 (top panels), pMIG-ICN.1 and pMIT-Bcl-2 (second row panels), or pMIG-ICN.1 and pMIG-Bcl-2 (third row panels), or pMIG-hTERT and pMIT-Bcl-2 (bottom panels). The cells were maintained in DMEM supplemented with 15% fetal calf serum, and a cocktail of IL-6, IL-3 and SCF. Shown is the phenotype of representative clones 3 (young) months after 90 days of continuous of culture. The panels represent the results of the flow cytometric analysis for expression of the viral expression markers (GFP and Thy1.1), as well as four markers required to define long-term HSCs in mice, Sca-1, c-kit, CD34 and Flk-2. The four cell lines contained subpopulations that retained the phenotypes of lt-HSCs (Sca-1+, c-kit+, CD34-, flk-2-).

[0172] Referring to FIG. 11, this figure shows the phenotypic comparison of cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with different combinations of oncogenes and maintained in continuous in vitro culture for >90 days. 5FU enriched HSCs were retroviral transduced with pMIG-MYC and pMIG-ICN.1 (top panels), pMIG-MYC.ER and pMIG-ICN.1 (middle panels), or pMIG-ICN.1.ER and pMIG-MYC. The cells were maintained in DMEM supplemented with 15% fetal calf serum, and a cocktail of IL-6, IL-3 and SCF. Shown is the phenotype of representative clones 3 (young) months after 90 days of continuous of culture. The panels represent the results of the flow cytometric analysis for expression of the viral expression markers (GFP and Thy1.1), as well as four markers required to define long-term HSCs in mice, Sca-1, c-kit, CD34 and Flk-2. The four cell lines contained subpopulations that retained the phenotypes of lt-HSCs (Sca-1+, c-kit+, CD34-, flk-2-).

[0173] These cell lines have also been used to reconstitute cellular compartments in vivo. Referring to FIG. 12, this figures shows the results of in vivo reconstitution of T cell and B cell compartments from cell lines derived from HSCs obtained from young C57/BL6 mice that were retrovirally transduced with different combinations of oncogenes and maintained in continuous in vitro culture for >90 days. Briefly, 5FU enriched HSCs were retroviral transduced with pMIG-ICN.1-ER and pMIG-hTERT (top panels), pMIG-MYC.ER and pMIG-hTERT (middle panels), or pMIG-MYC-ER and pMIT-Bcl-2 (lower panels). The cell lines were maintained in DMEM supplemented with 15% fetal calf serum, and a cocktail of IL-6, IL-3 and SCF. Lethally irradiated young C57/BL6 mice were reconstituted using bone marrow stem cells from Rag2-/- mice and LT-HSC lines generated in vitro. Six weeks later, bone marrow was harvested and stained with a panel of specific lineage markers. The development of mature CD4 and B220 positive/GFP positive cells can readily be observed. Data from four representative mice are presented in this figure. In each group, approximately 30% of the mice retain GFP marker.

Example 4

[0174] The following example describes an extension of the method for reversibly immortalizing human cord blood and bone marrow derived HSCs in vitro.

[0175] One additional application of this technology is the ability to expand human long-term hematopoietic stem cells in vitro through their conditional immortalization. The inventors have therefore adapted the in vitro method described in the previous examples for human cells with a few changes. First, the retroviruses are packaged preferably with amphotrophic envelopes in order to enable efficient transduction of human cells. In addition, the source of the cells is human cord blood obtained anonymously from the a cord blood bank, following all rules and regulations set forth by the Institutional Review Boards of the inventors' institutions. The resulting cells will express reporter genes that may ultimately be useful for isolating a pure population by high speed cell sorting. The inventors have noticed that many mature cells resulting from the murine lt-HSC cell lines lose expression of the surface markers, potentially due to the methylation of the retroviral genome upon lineage determination and differentiation. The inventors expect to see similar behavior in the human cells, in which case the lt-HSCs and their prevalence in transplant recipients can be monitored by the presence of reporter genes in such cells, in combination with cell surface markers for that population of cells.

Example 5

[0176] The following example describes an approach to the sequential excision of the DNA fragments encoding MYC-ER and Bcl-2 from conditionally immortalized HSC cells.

[0177] In order to avoid taking the risk of introducing HSCs that harbor transgenes encoding MYC.ER and Bcl-2 into humans and/or mice, these two DNA fragments will be excised using a bacterial recombinase approach. Two different recombinases will be used in order to allow control over which one of the two genes is excised at any one point in time. Two examples of such recombinases are the Cre and Flp recombinases. Briefly, the recognition substrate sequences (RSS's) for one of the recombinases is introduced into the retroviral constructs such that they flank the open reading frame of the oncogene, as well as the reporter gene (GFP or Thy1.1). In this case, the cells are incubated in media containing a Tat-Cre fusion protein. This recombinant protein has been previously described and shown to be able to passively enter cells, and mediate loxP site-dependent recombination of genomic DNA.

[0178] This approach will allow the achievement of a number of things in order to enable the generation of many HSCs for differentiation in vitro and in vivo. First, the cells can gradually be weaned from the high levels of proliferative and survival signals they had become accustomed to during the conditional transformation process. Second, the cells can be re-adapted to depend on normal cytokines for their homeostatic functions and differentiation. Third, the sequential loss of reporter expression will allow the definition of the status and degree of deletion of each one of the genes in question. Accordingly, cells that express both reporter genes (GFP and Thy1.1) harbor both sequences (MYC and Bcl-2, respectively), cells that express Thy1.1 but no GFP have successfully deleted the MYC encoding sequences, but still contain Bcl-2 genes, and lastly, cells that do not express either GFP or Thy1.1 have deleted both of those alleles. FIG. 12 represents this approach in a diagram.

[0179] In addition, this approach is tested in mice by obtaining 5FU enriched BM-HSCs from a strain of mice in which the expression of a human MYC transgene can be induced by the withdrawal of tetracycline and the presence of a bacterial protein called tTA (tetracycline transactivator protein). The human MYC cDNA was cloned downstream of a tetracycline regulatory transcription element (TRE). The TRE-MYC mice are treated with 5FU and used to harvest BM-HSCs. Those cells are transduced in vitro with retroviruses expressing Bcl-2 and tTA (pMIT-Bcl2 and pMIG-tTA). The cells are cultured in the continuous presence of Doxycycline in order to maintain the MYC transgene silent. Once the cells are analyzed by flow cytometry, they can be used for transplantation back into mice that will not be maintained on a doxycycline containing diet (this is a more stable form of tetracycline is normally used in vivo).

[0180] Once the lt-HSC cell lines are generated, the effect of culturing them in the presence of doxycycline in vitro will be examined in parallel with MYC.ER harboring cell lines that will be cultured in the absence of 4OHT. The protein levels of MYC are monitored by western blots and intracellular staining approaches throughout.

Example 6

[0181] The following example describes the generation of many hematopoietic lineages in vitro, following the withdrawal of 4OHT from the liquid tissue culture media.

[0182] The traditional methods used to determine the potency of an HSC involve the use of semi-solid media (methycellulose) with defined cytokines in order to potentiate the differentiation of HSCs into specific lineages. The inventors were interested in determining the pluripotency of this cell population created using the method of the present invention in vitro. In order to examine this issue, the ABM42 and ABM46 cell lines described herein were maintained in media containing IL-3, IL-6 and SCF, but without 4OHT. In addition to the lineages that the inventors were able to detect in the reconstituted mice (i.e., lymphoid, myeloid and granulocytic), GFP+ cells could also be detected that expressed NK1.1 or ter-119 (FIG. 13). The NK1.1 cells could either be NK-cell, or NK-T cells. The ter-119 expressing cells are of the erythroid lineage. These findings indicate that these cell lines are capable of giving rise to all of the elements of a normal hematopoietic system and that the cells will be useful for generation of large quantities of specific elements to be used for passive therapies. In addition, they will be of great use and importance to study the early events in hematopoiesis and to identify novel therapy for therapeutic intervention in genetic disorders, or complications that arise the normal course of chemotherapy, or even infectious disease.

Example 7

[0183] The following example describes a method for high throughput screens of small molecules or biological agents that induce or inhibit differentiation in conditionally transformed long term HSCs.

[0184] The following is a general method for screening small molecules or biological agents that induce or inhibit HSC differentiation. Previously, these types of large screens were prohibited by the fact that large numbers of stem cells were unobtainable. With the present inventors current ability to conditionally immortalize long term HSCs, it is now feasible to propose such technologies.

[0185] By way of example, one such method is a myeloid differentiation read-out that has been adapted from Schneider, et al. (Schneider, T., and Issekutz, A. C. (1996). Quantitation of eosinophil and neutrophil infiltration into rat lung by basic assays for eosinophil peroxidase and myeloperoxidase. Application in a Brown Norway rat model of allergic pulmonary inflammation. J Immunol Methods 198, 1-14). Briefly, conditionally transformed long term HSCs are plated in 96 well, flat bottom plates at various concentrations of cell numbers (usually 2.times.10.sup.4-5.times.10.sup.4 cells/well). The screens are carried out either in complete media (DMEM+15% heat inactivated fetal calf serum, 1.times. penicillin/streptomycin, 1.times.1-glutamine and 1.times. non-essential amino acids, supplemented with IL-3, IL-6 and SCF) with added 4OHT in order to maintain the cells in an undifferentiated state, or in the absence of added 4OHT in order to induce differentiation. These conditions have been shown to give rise to Mac-1+ cells, consistent with a myeloid differentiation pattern. Additional cytokines can be added to direct differentiation in specific paths, although this system can also be used to screen for specific functions of a panel of cytokines. In this instance, the complete media will be added without supplementation with IL-3, IL-6 and SCF, but instead with the given cytokines to be tested or used to direct differentiation (e.g., CSF-1, G-CSF, GM-CSF, EPO, TEPO, etc.).

[0186] Small molecules, biological agents or positive control substances (e.g., Arsenic 0.sub.3) are titrated across the 96 well plate and incubated with the ltHSCs for time frames ranging from 24 to 72 hours, or longer, if needed and as determined based on the agents or molecules to be tested. After incubation, the cells are washed with PBS and resuspended in PBS for overnight storage at -80.degree. C. to lyse the cells. The cells are then thawed at room temperature and the plates are centrifuged for 10 min at 3,000 rpm. The supernatant is then transferred to a new 96 well plate and mixed with tetramethybenzidine (TMB) for 40 min. The reaction is stopped with 4N H.sub.2SO.sub.4 and the O.D. is read at 450 nm. This type of high-throughput assay can be used to test small molecules or biological agents for the ability to induce or block the differentiation of conditionally transformed long term HSCs into a wide variety of cell types. Results of these screens can then further be tested for the ability to induce or inhibit HSC differentiation in vivo. Variations on this assay format will be apparent to those of skill in the art and are encompassed by the present invention.

Example 8

[0187] The following example describes the use of the method of the invention to generate cell lines of an intermediate hematopoietic lineage.

[0188] The following protocol can be used to induce the development of cell lines representing intermediate stages of hematopoietic lineage development following transplantation of conditionally immortalized lt-HSC cell lines into lethally irradiated mice. First, 10.sup.3-10.sup.5 conditionally transformed lt-HSC cell lines generated according to the method of the invention are transferred into cohorts of lethally irradiated recipient mice. The transplants will also include 10.sup.5 Rag-1.sup.-/- cells as carriers in order to ensure the initial survival of the irradiated mice. The mice are treated with weekly injections of 1 mg tamoxifen, intraperitoneally, in order to immortalize partially differentiated cells derived from the conditionally transformed lt-HSC cell lines. Injections begin either 3 days-1 week after the initial transplant, or 8 weeks after the transplant, once the mice have been fully reconstituted by the conditionally transformed lt-HSC cell lines. Cells are collected from the spleen and bone marrow cells from mice three days after treatment with tamoxifen, or when they show clinical signs associated with leukemias. The cells are cultured in either the standard bone marrow culture conditions with 4-OHT (DMEM, 15% fetal calf serum, pen/strep, L-glut, non essential amino acids, IL-3, IL-6 and SCF), or in the presence of other cytokines and medium used for different hematopoietic cell types. Cell lines are frozen and/or expanded, and cell lines are also single-cell cloned by limiting dilution and defined by PCR amplification of proviral integrations, frozen, and then characterized for surface marker expression by flow cytometry. These types of approaches are used for both murine and human ctlt-HSC cell lines, using either NOD/SCID mice as the recipients, or neonatal Rag-1-/- mice, which will be given intrahepatic injections.

Example 9

[0189] The following example describes the use of the method of the invention and the adoption of protocols used to generate mature CD4+.alpha..beta. T-cells in vitro to develop cell lines representing intermediate stages of T-cell development.

[0190] In this experiment, conditionally immortalized lt-HSC cell lines generated according to the method of the invention are plated in the presence of the normal cytokine cocktail, supplemented with IL-7 and without tamoxifen. Parallel cultures are established on a layer of OP-9 stromal cells that express Jagged, a Notch-1 ligand. Cells are stained for T-cell lineage markers every 48 hours after the cultures are initiated to monitor for signs of T-cell development. The wells that show signs of T-lineage commitment and development are switched to media containing tamoxifen in order to stabilize the phenotype and establish cell lines. The resulting cell lines are expanded, cloned and characterized as described in Example 8. The T-cell lines are specifically stained for individual TCR-V.beta. alleles in order to determine their T-cell receptor repertoire usage. Some mature T-cell lines, or cell lines representing progenitor populations, are transplanted into Rag-1.sup.-/- mice in order to evaluate their ability to conform to normal tolerance and homeostatic mechanisms in vivo, as well as their ability to further differentiate in vivo, when appropriate. Finally, their ability to respond to antigenic stimulation is evaluated in vitro and in vivo.

Example 10

[0191] The following example describes the use of the method of the invention and the adoption of protocols used for the directed differentiation of HSCs into myeloid cell lineages to develop intermediate developmental cell lines and myeloid leukemia models.

[0192] In this experiment, conditionally immortalized lt-HSC cell lines generated according to the methods of the present invention are plated in the presence of the normal cytokine cocktail, supplemented with G-CSF and without tamoxifen. Cells are stained for myeloid lineage markers every 48 hours after the cultures are initiated to monitor for signs of myeloid development. The wells that show signs of myeloid lineage commitment and development are switched to media containing tamoxifen in order to stabilize the phenotype and establish cell lines. The resulting cell lines are expanded, cloned and characterized as described in Example 8. Some of the resulting cell lines are transplanted back into mice in order to monitor their ability to repopulate Op/Op mice (mutant mice that naturally lack macrophages). Those cell lines are also transplanted into wild type mice that will be maintained on tamoxifen throughout, in order to determine if these cell lines will also give rise to myeloid leukemias similar to human AML, CML and APL. These novel tumors provide novel models for preclinical therapeutics.

Example 11

[0193] The following example describes the generation of Human adult ctlt-HSC cell lines and examination of their pluripotential in vivo using NOD/SCID or RAG.sup.-/- xenotransplant models.

[0194] In this experiment, CD34+ cells (from mobilized blood or cord blood) are transduced in vitro with retroviral vectors encoding MYC-ER, Bcl-2 and GFP (for later detection of transplanted cells), packaged using amphotrophic envelopes (according to the methods of the present invention). lt-HSC are selected by propagation in vitro in the presence of 4OHT and growth factors, as described above using murine HSCs. Pluripotency of the selected cells is evaluated by transplantation of lt-HSC lines into sublethally irradiated NOD/SCID or NOD/SCID/.beta.-2M.sup.-/- or Rag-1.sup.-/- or Rag-2.sup.-/- mice, followed 6-12 weeks later by analysis of all blood cell lineages by immunofluorescence flow cytometry. More particularly, following the generation of ctlt-HSC cell lines using the method of the present invention, one can use two different and complimentary approaches to examine their pluripotency. In a first approach, varying amounts of clonal ctlt-HSC cell lines are introduced into sublethally irradiated NOD/SCID mice or NOD/SCID/.beta.-2M.sup.-/- mice. In this instance, 10.sup.3-10.sup.5 cells derived from a human ctlt-HSC cell lines are transferred intravenously after the mice are subjected to a sublethal irradiation regimen (0.3 Gy). The mice are analyzed for reconstitution at 6-12 weeks after transplantation. Second, 10.sup.3-10.sup.5 cells derived from a human ctlt-HSC cell lines are introduced into the liver of neonatal Rag-1.sup.-/- or Rag-2 mice by direct injection. Those xenotransplants will also be analyzed for appropriate reconstitution 6-12 weeks after transplantation.

Example 12

[0195] The following example describes the use of conditional approaches to abrogate expression of MYC and Bcl-2 from the ctlt-HSCs after transplantation.

[0196] In this experiment, viruses (viral vectors) used to transform stem cells are re-engineered to contain two loxP sites flanking the MYC-ER, Bcl-2 and GFP open reading frames (ORFS). When the cells are transplanted, a regulated form of Cre or CRE-TAT fusion protein will be used to delete the oncogene-encoding sequences, thus eliminating risk of insert-driven malignancy in recipients. This approach is first developed in mice, then applied to human lt-HSCs.

[0197] In a second approach, lt-HSCs from TRE-MYC mice are used to generate the cell lines with retroviruses that encode Bcl-2 or rtTA. These are transplanted into mice. Mice are fed Doxycycline to abrogate the expression of MYC. One can use lt-HSCs obtained from TRE-MYCxTRE-Bcl-2 bigenic mice that can be transduced with a pMIG-rtTA retrovirus to eliminate MYC and Bcl-2 expression.

Example 13

[0198] The following example describes the use of HIV-1 Tat protein fusions with MYC and/or Bcl-2 to attain conditional transformation without genetic modification of the lt-HSCs.

[0199] MYC-Tat and Bcl-2-Tat fusion proteins are generated and purified using established protocols. The fusion proteins are tested by treatment of cells in which one can easily assay the effects of overexpressed Bcl-2 (e.g., activated T cells, B-cell lymphoma cell lines that are rendered resistant to BCMA-Fc, etc.) or MYC (e.g., anergic B-cells, naive T-cells, activated T-cells). Combinations of MYC-Tat and Bcl-2-Tat proteins are used to allow propagation of lt-HSCs prior to transplantation. This approach is readily developed and tested in the mouse system, then applied to human.

[0200] The entire disclosure of each of U.S. Provisional patent Application No. 60/728,131 and U.S. provisional Patent Application 60/765,993 is incorporated herein by reference.

[0201] While various embodiments of the present invention have been described in detail, it is apparent that modifications and adaptations of those embodiments will occur to those skilled in the art. It is to be expressly understood, however, that such modifications and adaptations are within the scope of the present invention, as set forth in the following claims.

Sequence CWU 1

1

1412377DNAHomo sapiens 1acccccgagc tgtgctgctc gcggccgcca ccgccgggcc ccggccgtcc ctggctcccc 60tcctgcctcg agaagggcag ggcttctcag aggcttggcg ggaaaaagaa cggagggagg 120gatcgcgctg agtataaaag ccggttttcg gggctttatc taactcgctg tagtaattcc 180agcgagaggc agagggagcg agcgggcggc cggctagggt ggaagagccg ggcgagcaga 240gctgcgctgc gggcgtcctg ggaagggaga tccggagcga atagggggct tcgcctctgg 300cccagccctc ccgctgatcc cccagccagc ggtccgcaac ccttgccgca tccacgaaac 360tttgcccata gcagcgggcg ggcactttgc actggaactt acaacacccg agcaaggacg 420cgactctccc gacgcgggga ggctattctg cccatttggg gacacttccc cgccgctgcc 480aggacccgct tctctgaaag gctctccttg cagctgctta gacgctggat ttttttcggg 540tagtggaaaa ccagcagcct cccgcgacga tgcccctcaa cgttagcttc accaacagga 600actatgacct cgactacgac tcggtgcagc cgtatttcta ctgcgacgag gaggagaact 660tctaccagca gcagcagcag agcgagctgc agcccccggc gcccagcgag gatatctgga 720agaaattcga gctgctgccc accccgcccc tgtcccctag ccgccgctcc gggctctgct 780cgccctccta cgttgcggtc acacccttct cccttcgggg agacaacgac ggcggtggcg 840ggagcttctc cacggccgac cagctggaga tggtgaccga gctgctggga ggagacatgg 900tgaaccagag tttcatctgc gacccggacg acgagacctt catcaaaaac atcatcatcc 960aggactgtat gtggagcggc ttctcggccg ccgccaagct cgtctcagag aagctggcct 1020cctaccaggc tgcgcgcaaa gacagcggca gcccgaaccc cgcccgcggc cacagcgtct 1080gctccacctc cagcttgtac ctgcaggatc tgagcgccgc cgcctcagag tgcatcgacc 1140cctcggtggt cttcccctac cctctcaacg acagcagctc gcccaagtcc tgcgcctcgc 1200aagactccag cgccttctct ccgtcctcgg attctctgct ctcctcgacg gagtcctccc 1260cgcagggcag ccccgagccc ctggtgctcc atgaggagac accgcccacc accagcagcg 1320actctgagga ggaacaagaa gatgaggaag aaatcgatgt tgtttctgtg gaaaagaggc 1380aggctcctgg caaaaggtca gagtctggat caccttctgc tggaggccac agcaaacctc 1440ctcacagccc actggtcctc aagaggtgcc acgtctccac acatcagcac aactacgcag 1500cgcctccctc cactcggaag gactatcctg ctgccaagag ggtcaagttg gacagtgtca 1560gagtcctgag acagatcagc aacaaccgaa aatgcaccag ccccaggtcc tcggacaccg 1620aggagaatgt caagaggcga acacacaacg tcttggagcg ccagaggagg aacgagctaa 1680aacggagctt ttttgccctg cgtgaccaga tcccggagtt ggaaaacaat gaaaaggccc 1740ccaaggtagt tatccttaaa aaagccacag catacatcct gtccgtccaa gcagaggagc 1800aaaagctcat ttctgaagag gacttgttgc ggaaacgacg agaacagttg aaacacaaac 1860ttgaacagct acggaactct tgtgcgtaag gaaaagtaag gaaaacgatt ccttctaaca 1920gaaatgtcct gagcaatcac ctatgaactt gtttcaaatg catgatcaaa tgcaacctca 1980caaccttggc tgagtcttga gactgaaaga tttagccata atgtaaactg cctcaaattg 2040gactttgggc ataaaagaac ttttttatgc ttaccatctt ttttttttct ttaacagatt 2100tgtatttaag aattgttttt aaaaaatttt aagatttaca caatgtttct ctgtaaatat 2160tgccattaaa tgtaaataac tttaataaaa cgtttatagc agttacacag aatttcaatc 2220ctagtatata gtacctagta ttataggtac tataaaccct aatttttttt atttaagtac 2280attttgcttt ttaaagttga tttttttcta ttgtttttag aaaaaataaa ataactggca 2340aatatatcat tgagccaaaa aaaaaaaaaa aaaaaaa 23772454PRTHomo sapiens 2Met Asp Phe Phe Arg Val Val Glu Asn Gln Gln Pro Pro Ala Thr Met 1 5 10 15 Pro Leu Asn Val Ser Phe Thr Asn Arg Asn Tyr Asp Leu Asp Tyr Asp 20 25 30 Ser Val Gln Pro Tyr Phe Tyr Cys Asp Glu Glu Glu Asn Phe Tyr Gln 35 40 45 Gln Gln Gln Gln Ser Glu Leu Gln Pro Pro Ala Pro Ser Glu Asp Ile 50 55 60 Trp Lys Lys Phe Glu Leu Leu Pro Thr Pro Pro Leu Ser Pro Ser Arg 65 70 75 80 Arg Ser Gly Leu Cys Ser Pro Ser Tyr Val Ala Val Thr Pro Phe Ser 85 90 95 Leu Arg Gly Asp Asn Asp Gly Gly Gly Gly Ser Phe Ser Thr Ala Asp 100 105 110 Gln Leu Glu Met Val Thr Glu Leu Leu Gly Gly Asp Met Val Asn Gln 115 120 125 Ser Phe Ile Cys Asp Pro Asp Asp Glu Thr Phe Ile Lys Asn Ile Ile 130 135 140 Ile Gln Asp Cys Met Trp Ser Gly Phe Ser Ala Ala Ala Lys Leu Val 145 150 155 160 Ser Glu Lys Leu Ala Ser Tyr Gln Ala Ala Arg Lys Asp Ser Gly Ser 165 170 175 Pro Asn Pro Ala Arg Gly His Ser Val Cys Ser Thr Ser Ser Leu Tyr 180 185 190 Leu Gln Asp Leu Ser Ala Ala Ala Ser Glu Cys Ile Asp Pro Ser Val 195 200 205 Val Phe Pro Tyr Pro Leu Asn Asp Ser Ser Ser Pro Lys Ser Cys Ala 210 215 220 Ser Gln Asp Ser Ser Ala Phe Ser Pro Ser Ser Asp Ser Leu Leu Ser 225 230 235 240 Ser Thr Glu Ser Ser Pro Gln Gly Ser Pro Glu Pro Leu Val Leu His 245 250 255 Glu Glu Thr Pro Pro Thr Thr Ser Ser Asp Ser Glu Glu Glu Gln Glu 260 265 270 Asp Glu Glu Glu Ile Asp Val Val Ser Val Glu Lys Arg Gln Ala Pro 275 280 285 Gly Lys Arg Ser Glu Ser Gly Ser Pro Ser Ala Gly Gly His Ser Lys 290 295 300 Pro Pro His Ser Pro Leu Val Leu Lys Arg Cys His Val Ser Thr His 305 310 315 320 Gln His Asn Tyr Ala Ala Pro Pro Ser Thr Arg Lys Asp Tyr Pro Ala 325 330 335 Ala Lys Arg Val Lys Leu Asp Ser Val Arg Val Leu Arg Gln Ile Ser 340 345 350 Asn Asn Arg Lys Cys Thr Ser Pro Arg Ser Ser Asp Thr Glu Glu Asn 355 360 365 Val Lys Arg Arg Thr His Asn Val Leu Glu Arg Gln Arg Arg Asn Glu 370 375 380 Leu Lys Arg Ser Phe Phe Ala Leu Arg Asp Gln Ile Pro Glu Leu Glu 385 390 395 400 Asn Asn Glu Lys Ala Pro Lys Val Val Ile Leu Lys Lys Ala Thr Ala 405 410 415 Tyr Ile Leu Ser Val Gln Ala Glu Glu Gln Lys Leu Ile Ser Glu Glu 420 425 430 Asp Leu Leu Arg Lys Arg Arg Glu Gln Leu Lys His Lys Leu Glu Gln 435 440 445 Leu Arg Asn Ser Cys Ala 450 351552DNAHomo sapiensmodified_base(31450)..(31450)a, c, g or t 3acttgagccc aagagttcaa ggctacggtg agccatgatt gcaacaccac acgccagcct 60tggtgacaga atgagaccct gtctcaaaaa aaaaaaaaaa aattgaaata atataaagca 120tcttctctgg ccacagtgga acaaaaccag aaatcaacaa caagaggaat tttgaaaact 180atacaaacac atgaaaatta aacaatatac ttctgaatga ccagtgagtc aatgaagaaa 240ttaaaaagga aattgaaaaa tttatttaag caaatgataa cggaaacata acctctcaaa 300acccacggta tacagcaaaa gcagtgctaa gaaggaagtt tatagctata agcagctaca 360tcaaaaaagt agaaaagcca ggcgcagtgg ctcatgcctg taatcccagc actttgggag 420gccaaggcgg gcagatcgcc tgaggtcagg agttcgagac cagcctgacc aacacagaga 480aaccttgtcg ctactaaaaa tacaaaatta gctgggcatg gtggcacatg cctgtaatcc 540cagctactcg ggaggctgag gcaggataac cgcttgaacc caggaggtgg aggttgcggt 600gagccgggat tgcgccattg gactccagcc tgggtaacaa gagtgaaacc ctgtctcaag 660aaaaaaaaaa aagtagaaaa acttaaaaat acaacctaat gatgcacctt aaagaactag 720aaaagcaaga gcaaactaaa cctaaaattg gtaaaagaaa agaaataata aagatcagag 780cagaaataaa tgaaactgaa agataacaat acaaaagatc aacaaaatta aaagttggtt 840ttttgaaaag ataaacaaaa ttgacaaacc tttgcccaga ctaagaaaaa aggaaagaag 900acctaaataa ataaagtcag agatgaaaaa agagacatta caactgatac cacagaaatt 960caaaggatca ctagaggcta ctatgagcaa ctgtacacta ataaattgaa aaacctagaa 1020aaaatagata aattcctaga tgcatacaac ctaccaagat tgaaccatga agaaatccaa 1080agcccaaaca gaccaataac aataatggga ttaaagccat aataaaaagt ctcctagcaa 1140agagaagccc aggacccaat ggcttccctg ctggatttta ccaatcattt aaagaagaat 1200gaattccaat cctactcaaa ctattctgaa aaatagagga aagaatactt ccaaactcat 1260tctacatggc cagtattacc ctgattccaa aaccagacaa aaacacatca aaaacaaaca 1320aacaaaaaaa cagaaagaaa gaaaactaca ggccaatatc cctgatgaat actgatacaa 1380aaatcctcaa caaaacacta gcaaaccaaa ttaaacaaca ccttcgaaag atcattcatt 1440gtgatcaagt gggatttatt ccagggatgg aaggatggtt caacatatgc aaatcaatca 1500atgtgataca tcatcccaac aaaatgaagt acaaaaacta tatgattatt tcactttatg 1560cagaaaaagc atttgataaa attctgcacc cttcatgata aaaaccctca aaaaaccagg 1620tatacaagaa acatacaggc caggcacagt ggctcacacc tgcgatccca gcactctggg 1680aggccaaggt gggatgattg cttgggccca ggagtttgag actagcctgg gcaacaaaat 1740gagacctggt ctacaaaaaa cttttttaaa aaattagcca ggcatgatgg catatgcctg 1800tagtcccagc tagtctggag gctgaggtgg gagaatcact taagcctagg aggtcgaggc 1860tgcagtgagc catgaacatg tcactgtact ccagcctaga caacagaaca agaccccact 1920gaataagaag aaggagaagg agaagggaga agggagggag aagggaggag gaggagaagg 1980aggaggtgga ggagaagtgg aaggggaagg ggaagggaaa gaggaagaag aagaaacata 2040tttcaacata ataaaagccc tatatgacag accgaggtag tattatgagg aaaaactgaa 2100agcctttcct ctaagatctg gaaaatgaca agggcccact ttcaccactg tgattcaaca 2160tagtactaga agtcctagct agagcaatca gataagagaa agaaataaaa ggcatccaaa 2220ctggaaagga agaagtcaaa ttatcctgtt tgcagatgat atgatcttat atctggaaaa 2280gacttaagac accactaaaa aactattaga gctgaaattt ggtacagcag gatacaaaat 2340caatgtacaa aaatcagtag tatttctata ttccaacagc aaacaatctg aaaaagaaac 2400caaaaaagca gctacaaata aaattaaaca gctaggaatt aaccaaagaa gtgaaagatc 2460tctacaatga aaactataaa atgttgataa aagaaattga agagggcaca aaaaaagaaa 2520agatattcca tgttcataga ttggaagaat aaatactgtt aaaatgtcca tactacccaa 2580agcaatttac aaattcaatg caatccctat taaaatacta atgacgttct tcacagaaat 2640agaagaaaca attctaagat ttgtacagaa ccacaaaaga cccagaatag ccaaagctat 2700cctgaccaaa aagaacaaaa ctggaagcat cacattacct gacttcaaat tatactacaa 2760agctatagta acccaaacta catggtactg gcataaaaac agatgagaca tggaccagag 2820gaacagaata gagaatccag aaacaaatcc atgcatctac agtgaactca tttttgacaa 2880aggtgccaag aacatacttt ggggaaaaga taatctcttc aataaatggt gctggaggaa 2940ctggatatcc atatgcaaaa taacaatact agaactctgt ctctcaccat atacaaaagc 3000aaatcaaaat ggatgaaagg cttaaatcta aaacctcaaa ctttgcaact actaaaagaa 3060aacaccggag aaactctcca ggacattgga gtgggcaaag acttcttgag taattccctg 3120caggcacagg caaccaaagc aaaaacagac aaatgggatc atatcaagtt aaaaagcttc 3180tgcccagcaa aggaaacaat caacaaagag aagagacaac ccacagaatg ggagaatata 3240tttgcaaact attcatctaa caaggaatta ataaccagta tatataagga gctcaaacta 3300ctctataaga aaaacaccta ataagctgat tttcaaaaat aagcaaaaga tctgggtaga 3360catttctcaa aataagtcat acaaatggca aacaggcatc tgaaaatgtg ctcaacacca 3420ctgatcatca gagaaatgca aatcaaaact actatgagag atcatctcat cccagttaaa 3480atggctttta ttcaaaagac aggcaataac aaatgccagt gaggatgtgg ataaaaggaa 3540acccttggac actgttggtg ggaatggaaa ttgctaccac tatggagaac agtttgaaag 3600ttcctcaaaa aactaaaaat aaagctacca tacagcaatc ccattgctag gtatatactc 3660caaaaaaggg aatcagtgta tcaacaagct atctccactc ccacatttac tgcagcactg 3720ttcatagcag ccaaggtttg gaagcaacct cagtgtccat caacagacga atggaaaaag 3780aaaatgtggt gcacatacac aatggagtac tacgcagcca taaaaaagaa tgagatcctg 3840tcagttgcaa cagcatgggg ggcactggtc agtatgttaa gtgaaataag ccaggcacag 3900aaagacaaac ttttcatgtt ctcccttact tgtgggagca aaaattaaaa caattgacat 3960agaaatagag gagaatggtg gttctagagg ggtgggggac agggtgacta gagtcaacaa 4020taatttattg tatgttttaa aataactaaa agagtataat tgggttgttt gtaacacaaa 4080gaaaggataa atgcttgaag gtgacagata ccccatttac cctgatgtga ttattacaca 4140ttgtatgcct gtatcaaaat atctcatgta tgctatagat ataaacccta ctatattaaa 4200aattaaaatt ttaatggcca ggcacggtgg ctcatgtccg taatcccagc actttgggag 4260gccgaggcgg gtggatcacc tgaggtcagg agtttgaaac cagtctggcc accatgatga 4320aaccctgtct ctactaaaga tacaaaaatt agccaggcgt ggtggcacat acctgtagtc 4380ccaactactc aggaggctga gacaggagaa ttgcttgaac ctgggaggcg gaggttgcag 4440tgagccgaga tcatgccact gcactgcagc ctgggtgaca gagcaagact ccatctcaaa 4500acaaaaacaa aaaaaagaag attaaaattg taatttttat gtaccgtata aatatatact 4560ctactatatt agaagttaaa aattaaaaca attataaaag gtaattaacc acttaatcta 4620aaataagaac aatgtatgtg gggtttctag cttctgaaga agtaaaagtt atggccacga 4680tggcagaaat gtgaggaggg aacagtggaa gttactgttg ttagacgctc atactctctg 4740taagtgactt aattttaacc aaagacaggc tgggagaagt taaagaggca ttctataagc 4800cctaaaacaa ctgctaataa tggtgaaagg taatctctat taattaccaa taattacaga 4860tatctctaaa atcgagctgc agaattggca cgtctgatca caccgtcctc tcattcacgg 4920tgcttttttt cttgtgtgct tggagatttt cgattgtgtg ttcgtgtttg gttaaactta 4980atctgtatga atcctgaaac gaaaaatggt ggtgatttcc tccagaagaa ttagagtacc 5040tggcaggaag caggtggctc tgtggacctg agccacttca atcttcaagg gtctctggcc 5100aagacccagg tgcaaggcag aggcctgatg acccgaggac aggaaagctc ggatgggaag 5160gggcgatgag aagcctgcct cgttggtgag cagcgcatga agtgccctta tttacgcttt 5220gcaaagattg ctctggatac catctggaaa aggcggccag cgggaatgca aggagtcaga 5280agcctcctgc tcaaacccag gccagcagct atggcgccca cccgggcgtg tgccagaggg 5340agaggagtca aggcacctcg aagtatggct taaatctttt tttcacctga agcagtgacc 5400aaggtgtatt ctgagggaag cttgagttag gtgccttctt taaaacagaa agtcatggaa 5460gcacccttct caagggaaaa ccagacgccc gctctgcggt catttacctc tttcctctct 5520ccctctcttg ccctcgcggt ttctgatcgg gacagagtga cccccgtgga gcttctccga 5580gcccgtgctg aggaccctct tgcaaagggc tccacagacc cccgccctgg agagaggagt 5640ctgagcctgg cttaataaca aactgggatg tggctggggg cggacagcga cggcgggatt 5700caaagactta attccatgag taaattcaac ctttccacat ccgaatggat ttggatttta 5760tcttaatatt ttcttaaatt tcatcaaata acattcagga ctgcagaaat ccaaaggcgt 5820aaaacaggaa ctgagctatg tttgccaagg tccaaggact taataaccat gttcagaggg 5880atttttcgcc ctaagtactt tttattggtt ttcataaggt ggcttagggt gcaagggaaa 5940gtacacgagg agaggcctgg gcggcagggc tatgagcacg gcagggccac cggggagaga 6000gtccccggcc tgggaggctg acagcaggac cactgaccgt cctccctggg agctgccaca 6060ttgggcaacg cgaaggcggc cacgctgcgt gtgactcagg accccatacc ggcttcctgg 6120gcccacccac actaacccag gaagtcacgg agctctgaac ccgtggaaac gaacatgacc 6180cttgcctgcc tgcttccctg ggtgggtcaa gggtaatgaa gtggtgtgca ggaaatggcc 6240atgtaaatta cacgactctg ctgatgggga ccgttccttc catcattatt catcttcacc 6300cccaaggact gaatgattcc agcaacttct tcgggtgtga caagccatga caaaactcag 6360tacaaacacc actcttttac taggcccaca gagcacggsc cacacccctg atatattaag 6420agtccaggag agatgaggct gctttcagcc accaggctgg ggtgacaaca gcggctgaac 6480agtctgttcc tctagactag tagaccctgg caggcactcc cccagattct agggcctggt 6540tgctgcttcc cgagggcgcc atctgccctg gagactcagc ctggggtgcc acactgaggc 6600cagccctgtc tccacaccct ccgcctccag gcctcagctt ctccagcagc ttcctaaacc 6660ctgggtgggc cgtgttccag cgctactgtc tcacctgtcc cactgtgtct tgtctcagcg 6720acgtagctcg cacggttcct cctcacatgg ggtgtctgtc tccttcccca acactcacat 6780gcgttgaagg gaggagattc tgcgcctccc agactggctc ctctgagcct gaacctggct 6840cgtggccccc gatgcaggtt cctggcgtcc ggctgcacgc tgacctccat ttccaggcgc 6900tccccgtctc ctgtcatctg ccggggcctg ccggtgtgtt cttctgtttc tgtgctcctt 6960tccacgtcca gctgcgtgtg tctctgcccg ctagggtctc ggggttttta taggcatagg 7020acgggggcgt ggtgggccag ggcgctcttg ggaaatgcaa catttgggtg tgaaagtagg 7080agtgcctgtc ctcacctagg tccacgggca caggcctggg gatggagccc ccgccaggga 7140cccgcccttc tctgcccagc actttcctgc ccccctccct ctggaacaca gagtggcagt 7200ttccacaagc actaagcatc ctcttcccaa aagacccagc attggcaccc ctggacattt 7260gccccacagc cctgggaatt cacgtgacta cgcacatcat gtacacactc ccgtccacga 7320ccgacccccg ctgttttatt ttaatagcta caaagcaggg aaatccctgc taaaatgtcc 7380tttaacaaac tggttaaaca aacgggtcca tccgcacggt ggacagttcc tcacagtgaa 7440gaggaacatg ccgtttataa agcctgcagg catctcaagg gaattacgct gagtcaaaac 7500tgccacctcc atgggatacg tacgcaacat gctcaaaaag aaagaatttc accccatggc 7560aggggagtgg ttaggggggt taaggacggt gggggcggca gctgggggct actgcacgca 7620ccttttacta aagccagttt cctggttctg atggtattgg ctcagttatg ggagactaac 7680cataggggag tggggatggg ggaacccgga ggctgtgcca tctttgccat gcccgagtgt 7740cctgggcagg ataatgctct agagatgccc acgtcctgat tcccccaaac ctgtggacag 7800aacccgcccg gccccagggc ctttgcaggt gtgatctccg tgaggaccct gaggtctggg 7860atccttcggg actacctgca ggcccgaaaa gtaatccagg ggttctggga agaggcgggc 7920aggagggtca gaggggggca gcctcaggac gatggaggca gtcagtctga ggctgaaaag 7980ggagggaggg cctcgagccc aggcctgcaa gcgcctccag aagctggaaa aagcggggaa 8040gggaccctcc acggagcctg cagcaggaag gcacggctgg cccttagccc accagggccc 8100atcgtggacc tccggcctcc gtgccatagg agggcactcg cgctgccctt ctagcatgaa 8160gtgtgtgggg atttgcagaa gcaacaggaa acccatgcac tgtgaatcta ggattatttc 8220aaaacaaagg tttacagaaa catccaagga cagggctgaa gtgcctccgg gcaagggcag 8280ggcaggcacg agtgatttta tttagctatt ttattttatt tacttacttt ctgagacaga 8340gttatgctct tgttgcccag gctggagtgc agcggcatga tcttggctca ctgcaacctc 8400cgtctcctgg gttcaagcaa ttctcgtgcc tcagcctccc aagtagctgg gatttcaggc 8460gtgcaccacc acacccggct aattttgtat ttttagtaga gatgggcttt caccatgttg 8520gtcaagctga tctcaaaatc ctgacctcag gtgatccgcc cacctcagcc tcccaaagtg 8580ctgggattac aggcatgagc cactgcacct ggcctattta accattttaa aacttccctg 8640ggctcaagtc acacccactg gtaaggagtt catggagttc aatttcccct ttactcagga 8700gttaccctcc tttgatattt tctgtaattc ttcgtagact ggggatacac cgtctcttga 8760catattcaca gtttctgtga ccacctgtta tcccatggga cccactgcag gggcagctgg 8820gaggctgcag gcttcaggtc ccagtggggt tgccatctgc cagtagaaac ctgatgtaga 8880atcagggcgc aagtgtggac actgtcctga atctcaatgt ctcagtgtgt gctgaaacat 8940gtagaaatta aagtccatcc ctcctactct actgggattg agccccttcc ctatcccccc 9000ccaggggcag aggagttcct ctcactcctg tggaggaagg aatgatactt tgttattttt 9060cactgctggt actgaatcca ctgtttcatt tgttggtttg tttgttttgt tttgagaggc 9120ggtttcactc ttgttgctca ggctggaggg agtgcaatgg cgcgatcttg gcttactgca 9180gcctctgcct cccaggttca agtgattctc ctgcttccgc ctcccatttg gctgggatta 9240caggcacccg ccaccatgcc cagctaattt tttgtatttt tagtagagac gggggtgggt 9300ggggttcacc atgttggcca ggctggtctc gaacttctga cctcagatga tccacctgcc 9360tctgcctcct aaagtgctgg gattacaggt gtgagccacc atgcccagct cagaatttac 9420tctgtttaga aacatctggg tctgaggtag gaagctcacc ccactcaagt gttgtggtgt 9480tttaagccaa tgatagaatt tttttattgt tgttagaaca ctcttgatgt tttacactgt 9540gatgactaag acatcatcag cttttcaaag acacactaac tgcacccata atactggggt

9600gtcttctggg tatcagcaat cttcattgaa tgccgggagg cgtttcctcg ccatgcacat 9660ggtgttaatt actccagcat aatcttctgc ttccatttct tctcttccct cttttaaaat 9720tgtgttttct atgttggctt ctctgcagag aaccagtgta agctacaact taacttttgt 9780tggaacaaat tttccaaacc gcccctttgc cctagtggca gagacaattc acaaacacag 9840ccctttaaaa aggcttaggg atcactaagg ggatttctag aagagcgacc tgtaatccta 9900agtatttaca agacgaggct aacctccagc gagcgtgaca gcccagggag ggtgcgaggc 9960ctgttcaaat gctagctcca taaataaagc aatttcctcc ggcagtttct gaaagtagga 10020aaggttacat ttaaggttgc gtttgttagc atttcagtgt ttgccgacct cagctacagc 10080atccctgcaa ggcctcggga gacccagaag tttctcgccc ccttagatcc aaacttgagc 10140aacccggagt ctggattcct gggaagtcct cagctgtcct gcggttgtgc cggggcccca 10200ggtctggagg ggaccagtgg ccgtgtggct tctactgctg ggctggaagt cgggcctcct 10260agctctgcag tccgaggctt ggagccaggt gcctggaccc cgaggctgcc ctccaccctg 10320tgcgggcggg atgtgaccag atgttggcct catctgccag acagagtgcc ggggcccagg 10380gtcaaggccg ttgtggctgg tgtgaggcgc ccggtgcgcg gccagcagga gcgcctggct 10440ccatttccca ccctttctcg acgggaccgc cccggtgggt gattaacaga tttggggtgg 10500tttgctcatg gtggggaccc ctcgccgcct gagaacctgc aaagagaaat gacgggcctg 10560tgtcaaggag cccaagtcgc ggggaagtgt tgcagggagg cactccggga ggtcccgcgt 10620gcccgtccag ggagcaatgc gtcctcgggt tcgtccccag ccgcgtctac gcgcctccgt 10680cctccccttc acgtccggca ttcgtggtgc ccggagcccg acgccccgcg tccggacctg 10740gaggcagccc tgggtctccg gatcaggcca gcggccaaag ggtcgccgca cgcacctgtt 10800cccagggcct ccacatcatg gcccctccct cgggttaccc cacagcctag gccgattcga 10860cctctctccg ctggggccct cgctggcgtc cctgcaccct gggagcgcga gcggcgcgcg 10920ggcggggaag cgcggcccag acccccgggt ccgcccggag cagctgcgct gtcggggcca 10980ggccgggctc ccagtggatt cgcgggcaca gacgcccagg accgcgctcc ccacgtggcg 11040gagggactgg ggacccgggc acccgtcctg ccccttcacc ttccagctcc gcctcctccg 11100cgcggacccc gccccgtccc gacccctccc gggtccccgg cccagccccc tccgggccct 11160cccagcccct ccccttcctt tccgcggccc cgccctctcc tcgcggcgcg agtttcaggc 11220agcgctgcgt cctgctgcgc acgtgggaag ccctggcccc ggccaccccc gcgatgccgc 11280gcgctccccg ctgccgagcc gtgcgctccc tgctgcgcag ccactaccgc gaggtgctgc 11340cgctggccac gttcgtgcgg cgcctggggc cccagggctg gcggctggtg cagcgcgggg 11400acccggcggc tttccgcgcg ctggtggccc agtgcctggt gtgcgtgccc tgggacgcac 11460ggccgccccc cgccgccccc tccttccgcc aggtgggcct ccccggggtc ggcgtccggc 11520tggggttgag ggcggccggg gggaaccagc gacatgcgga gagcagcgca ggcgactcag 11580ggcgcttccc ccgcaggtgt cctgcctgaa ggagctggtg gcccgagtgc tgcagaggct 11640gtgcgagcgc ggcgcgaaga acgtgctggc cttcggcttc gcgctgctgg acggggcccg 11700cgggggcccc cccgaggcct tcaccaccag cgtgcgcagc tacctgccca acacggtgac 11760cgacgcactg cgggggagcg gggcgtgggg gctgctgctg cgccgcgtgg gcgacgacgt 11820gctggttcac ctgctggcac gctgcgcgct ctttgtgctg gtggctccca gctgcgccta 11880ccaggtgtgc gggccgccgc tgtaccagct cggcgctgcc actcaggccc ggcccccgcc 11940acacgctagt ggaccccgaa ggcgtctggg atgcgaacgg gcctggaacc atagcgtcag 12000ggaggccggg gtccccctgg gcctgccagc cccgggtgcg aggaggcgcg ggggcagtgc 12060cagccgaagt ctgccgttgc ccaagaggcc caggcgtggc gctgcccctg agccggagcg 12120gacgcccgtt gggcaggggt cctgggccca cccgggcagg acgcgtggac cgagtgaccg 12180tggtttctgt gtggtgtcac ctgccagacc cgccgaagaa gccacctctt tggagggtgc 12240gctctctggc acgcgccact cccacccatc cgtgggccgc cagcaccacg caggcccccc 12300atccacatcg cggccaccac gtccctggga cacgccttgt cccccggtgt acgccgagac 12360caagcacttc ctctactcct caggcgacaa ggagcagctg cggccctcct tcctactcag 12420ctctctgagg cccagcctga ctggcgctcg gaggctcgtg gagaccatct ttctgggttc 12480caggccctgg atgccaggga ctccccgcag gttgccccgc ctgccccagc gctactggca 12540aatgcggccc ctgtttctgg agctgcttgg gaaccacgcg cagtgcccct acggggtgct 12600cctcaagacg cactgcccgc tgcgagctgc ggtcacccca gcagccggtg tctgtgcccg 12660ggagaagccc cagggctctg tggcggcccc cgaggaggag gacacagacc cccgtcgcct 12720ggtgcagctg ctccgccagc acagcagccc ctggcaggtg tacggcttcg tgcgggcctg 12780cctgcgccgg ctggtgcccc caggcctctg gggctccagg cacaacgaac gccgcttcct 12840caggaacacc aagaagttca tctccctggg gaagcatgcc aagctctcgc tgcaggagct 12900gacgtggaag atgagcgtgc gggactgcgc ttggctgcgc aggagcccag gtgaggaggt 12960ggtggccgtc gagggcccag gccccagagc tgaatgcagt aggggctcag aaaagggggc 13020aggcagagcc ctggtcctcc tgtctccatc gtcacgtggg cacacgtggc ttttcgctca 13080ggacgtcgag tggacacggt gatctctgcc tctgctctcc ctcctgtcca gtttgcataa 13140acttacgagg ttcaccttca cgttttgatg gacacgcggt ttccaggcgc cgaggccaga 13200gcagtgaaca gaggaggctg ggcgcggcag tggagccggg ttgccggcaa tggggagaag 13260tgtctggaag cacagacgct ctggcgaggg tgcctgcagg ttacctataa tcctcttcgc 13320aatttcaagg gtgggaatga gaggtgggga cgagaacccc ctcttcctgg gggtgggagg 13380taagggtttt gcaggtgcac gtggtcagcc aatatgcagg tttgtgttta agatttaatt 13440gtgtgttgac ggccaggtgc ggtggctcac gccggtaatc ccagcacttt gggaagctga 13500ggcaggtgga tcacctgagg tcaggagttt gagaccagcc tgaccaacat ggtgaaaccc 13560tatctgtact aaaaatacaa aaattagctg ggcatggtgg tgtgtgcctg taatcccagc 13620tacttgggag gctgaggcag gagaatcact tgaacccagg aggcggaggc tgcagtgagc 13680tgagattgtg ccattgtact ccagcctggg cgacaagagt gaaactctgt ctttaaaaaa 13740aaaaagtgtt cgttgattgt gccaggacag ggtagaggga gggagataag actgttctcc 13800agcacagatc ctggtcccat ctttaggtat gaagagggcc acatgggagc agaggacagc 13860agatggctcc acctgctgag gaagggacag tgtttgtggg tgttcagggg atggtgctgc 13920tgggccctgc cgtgtcccca ccctgttttt ctggatttga tgttgaggaa cctccgctcc 13980agcccccttt tggctcccag tgctcccagg ccctaccgtg gcagctagaa gaagtcccga 14040tttcaccccc tccccacaaa ctcccaagac atgtaagact tccggccatg cagacaagga 14100gggtgacctt cttggggctc ttttttttct ttttttcttt ttatggtggc aaaagtcata 14160taacatgaga ttggcactcc taacaccgtt ttctgtgtac agtgcagaat tgctaactcg 14220gcggtgttta cagcaggttg cttgaaatgc tgcgtcttgc gtgactggaa gtccctaccc 14280atcgaacggc agctgcctca cacctgctgc ggctcaggtg gaccacgccg agtcagataa 14340gcgtcatgca acccagtttt gctttttgtg ctccagcttc cttcgttgag gagagtttga 14400gttctctgat caggactctg cctgtcattg ctgttctctg acttcagatg aggtcacaat 14460ctgcccctgg cttatgcagg gagtgaggcg tggtccccgg gtgtccctgt cacgtgcagg 14520gtgagtgagg cgttgccccc aggtgtccct gtcacgtgta gggtgagtga ggcgcggccc 14580ccgggtgtcc ctgtcccgtg cagcgtgatt gaggtgtggc ccccgggtgt ccctgtcacg 14640tgtagggtga gtgaggcgcc atccccgggt gtccctgtca cgtgtagggt gagtgaggcg 14700tggtccccgg gtgtccctgt cccgtgcagg gtgagtgagg cactgtcccc gggtgtccct 14760gtcacgtgca gggtgagtga ggcgcggtcc ccgggtgtcc ctctcaggtg tagggtgagt 14820gaggcgcggc cccagggtgt ccctgtcacg tgtagggtga gtgaggcacc gtccctgggt 14880gtccctccca ggtatagggt gagtgaggca ctgtccccgg gtgtccctgt cacgtgcagg 14940gtgagtgagg cgcggccccc gggtgtccct ctcaggtgca gggtgagtga ggcgctgtcc 15000ctgggtgtcc ctgtctcgtg tagggtgagt gaggctctgt ccccaggtgt ccttggcgtt 15060tgctcacttg agcttgctcc tgaatgtttg ctctttctat agccacagct gcgccggttg 15120cccattgcct gggtagatgg tgcaggcgca gtgctggtcc ccaagcctat cttttctgat 15180gctcggctct tcttggtcac ctctccgttc cattttgcta cggggacacg ggactgcagg 15240ctctcgcctc ccgcgtgcca ggcactgcag ccacagcttc aggtccgctt gcctctgttg 15300ggcctggctt gctcaccacg tgcccgccac atgcatgctg ccaatactcc tctcccagct 15360tgtctcatgc cgaggctgga ctctgggctg cctgtgtctg ctgccacgtg ttgctggaga 15420catcccagaa agggttctct gtgccctgaa ggaaagcaag tcaccccagc cccctcactt 15480gtcctgtttt ctcccaagct gcccctctgc ttggccccct tgggtgggtg gcaacgcttg 15540tcaccttatt ctgggcacct gccgctcatt gcttaggctg ggctctgcct ccagtcgccc 15600cctcacatgg attgacgtcc agccacaggt tggagtgtct ctgtctgtct cctgctctga 15660gacccacgtg gagggccggt gtctccgcca gccttcgtca gacttccctc ttgggtctta 15720gttttgaatt tcactgattt acctctgacg tttctatctc tccattgtat gctttttctt 15780ggtttattct ttcattcctt ttctagcttc ttagtttagt catgcctttc cctctaagtg 15840ctgccttacc tgcaccctgt gttttgatgt gaagtaatct caacatcagc cactttcaag 15900tgttcttaaa atacttcaaa gtgttaatac ttcttttaag tattcttatt ctgtgatttt 15960tttctttgtg cacgctgtgt tttgacgtga aatcattttg atatcagtga cttttaagta 16020ttctttagct tattctgtga tttctttgag cagtgagtta tttgaacact gtttatgttc 16080aagatatgta gagtatcaag atacgtagag tattttaagt tatcatttta ttattgattt 16140ctaactcagt tgtgtagtgg tctgtataat accaattatt tgaagtttgc ggagccttgc 16200tttgtgatct agtgtgtgca tggtttccag aactgtccat tgtaaatttg acatcctgtc 16260aatagtgggc atgcatgttc actatatcca gcttattaag gtccagtgca aagcttctgt 16320ctccttctag atgcatgaaa ttccaagaag gaggccatag tccctcacct gggggatggg 16380tctgttcatt tcttctcgtt tggtagcatt tatgtgaggc attgttaggt gcatgcacgt 16440ggtagaattt ttatcttcct gatgagtgaa tcttttggag acttctatgt ctctagtaat 16500ctagtaattc tttttttaaa ttgctcttag tactgccaca ctgggcttct tttgattagt 16560attttcctgc tgtgtctgtt ttctgccttt aatttatata tatatatata tttttttttt 16620ttttgagaca gagtcttggt ctgtcgccca gggtgagtgc agtggtgtga tcacaggtca 16680gtgtaacttt taccttctgg cctgagccgt cctctcacct cagcctcctg agtagctgga 16740actgcagaca cgcaccgcta cacctggcta atttttaaat tttttctgga gacagggtct 16800tgctgtgttg cccaggctgg tctcaaactc ttggactcaa gggatccatc tacctcggct 16860tcccaaagtg ctgaattaca ggcatgagcc accatgtctg gcctaatttt caacactttt 16920atattcttat agtgtgggta tgtcctgtta acagcatgta ggtgaatttc caatccagtc 16980tgacagtcgt tgtttaactg gataacctga tttattttca tttttttgtc actagagacc 17040cgcctggtgc actctgattc tccacttgcc tgttgcatgt cctcgttccc ttgtttctca 17100ccacctcttg ggttgccatg tgcgtttcct gccgagtgtg tgttgatcct ctcgttgcct 17160cctggtcact gggcatttgc ttttatttct ctttgcttag tgttaccccc tgatcttttt 17220attgtcgttg tttgcttttg tttattgaga cagtctcact ctgtcaccca ggctggagtg 17280taatggcaca atctcggctc actgcaacct ctgcctcctc ggttcaagca gttctcattc 17340ctcaacctca tgagtagctg ggattacagg cgcccaccac cacgcctggc taatttttgt 17400atttttagta gagataggct ttcaccatgt tggccaggct ggtctcaaac tcctgacctc 17460aagtgatctg cccgccttgg cctcccacag tgctgggatt acaggtgcaa gccaccgtgc 17520ccggcatacc ttgatctttt aaaatgaagt ctgaaacatt gctacccttg tcctgagcaa 17580taagaccctt agtgtatttt agctctggcc accccccagc ctgtgtgctg ttttccctgc 17640tgacttagtt ctatctcagg catcttgaca cccccacaag ctaagcatta ttaatattgt 17700tttccgtgtt gagtgtttct gtagctttgc ccccgccctg cttttcctcc tttgttcccc 17760gtctgtcttc tgtctcaggc ccgccgtctg gggtcccctt ccttgtcctt tgcgtggttc 17820ttctgtcttg ttattgctgg taaaccccag ctttacctgt gctggcctcc atggcatcta 17880gcgacgtccg gggacctctg cttatgatgc acagatgaag atgtggagac tcacgaggag 17940ggcggtcatc ttggcccgtg agtgtctgga gcaccacgtg gccagcgttc cttagccagt 18000gagtgacagc aacgtccgct cggcctgggt tcagcctgga aaaccccagg catgtcgggg 18060tctggtggct ccgcggtgtc gagtttgaaa tcgcgcaaac ctgcggtgtg gcgccagctc 18120tgacggtgct gcctggcggg ggagtgtctg cttcctccct tctgcttggg aaccaggaca 18180aaggatgagg ctccgagccg ttgtcgccca acaggagcat gacgtgagcc atgtggataa 18240ttttaaaatt tctaggctgg gcgcggtggc tcacgcctgt aatcccagca ctttgggagg 18300ccaaggcggg tggatcacga ggtcaggagg tcgagaccat cctggccaac atgatgaaac 18360cccatctgta ctaaaaacac aaaaattagc tgggcgtggt ggcgggtgcc tgtaatccca 18420gctactcggg aggctgaggc aggagaattg cttgaacctg ggagttggaa gttgcagtga 18480gccgacattg caccactgca ctccagcctg gcaacacagc gagactctgt ctcaaaaaaa 18540aaaaaaaaaa aaaaaaaaaa aattctagta gccacattaa aaaagtaaaa aagaaaaggt 18600gaaattaatg taataataga ttttactgaa gcccagcatg tccacacctc atcattttag 18660ggtgttattg gtgggagcat cactcacagg acatttgaca ttttttgagc tttgtctgcg 18720ggatcccgtg tgtaggtccc gtgcgtggcc atctcggcct ggacctgctg ggcttcccat 18780ggccatggct gttgtaccag atggtgcagg tccgggatga ggtcgccagg ccctcagtga 18840gctggatgtg cagtgtccgg atggtgcacg tctgggatga ggtcgccagg ccctgctgtg 18900agctggatgt gtggtgtctg gatggtgcag gtcaggggtg aggtctccag gccctcggtg 18960agctggaggt atggagtccg gatgatgcag gtccggggtg aggtcgccag gccctgctgt 19020gagctggatg tgtggtgtct ggatggtgca ggtcaggggt gaggtctcca ggccctcggt 19080aagctggagg tatggagtcc ggatgatgca ggtccggggt gaggtcgcca ggccctgctg 19140tgagctggat gtgtggtgtc tggatggtgc aggtctgggg tgaggtcacc aggccctgcg 19200gtgagctggg tgtgcggtgt ctggatggtg caggtctgga gtgaggtcgc cagacggtgc 19260cagaccatgc ggtgagctgg atatgcggtg tccggatggt gcaggtctgg ggtgaggttg 19320ccaggccctg ctgtgagttg gatgtggggt gtccggatgc tgcaggtccg gtgtgaggtc 19380accaggccct gctgtgagct ggatgtgtgg tgtctggatg gtgcaggtct ggggtgaagg 19440tcgccaggcc cctgcttgtg agctggatgt gtggtgtctg gatggtgcag gtctggagtg 19500aggtcgccag gccctcggtg agctggatgt gcagtgtcca gatggtgcag gtccggggtg 19560aggtcgccag accctgcggt gagctggatg tgcggtgtct ggatggtgca ggtctggagt 19620gaggtcgcca ggccctcggt gagctggatg tatggagtcc ggatggtgcc ggtccggggt 19680gaggtcgcca gaccctgctg tgagctggat gtgcggtgtc tggatggtac aggtctggag 19740tgaggtcgcc agaccctgct gtgagctgga tatgcggtgt ccggatggtg caggtcaggg 19800gtgaggtctc caggccctcg gtgagctgga ggtatggagt ccggatgatg caggtccggg 19860gtgaggtcgc caggccctgc tgtgaactgg atgtgcggcg tctggatggt gcaggtctgg 19920ggtgtggtcg ccaggccctc ggtgagctgg aggtatggag tccggatgat gcaggtccgg 19980ggtgaggtcg ccaggccctg ctgtgagctg gatgtgcggc gtctggatgg tgcaggtctg 20040gggtgtggtc gccaggccct cggtgagctg gaggtatgga gtccggatga tgcaggtccg 20100gggtgaggtt gccaggccct gctgtgagct ggatgtgctg tatccggatg gtgcagtccg 20160gggtgaggtc gccaggccct gctgtgagct ggatgtgctg tatccggatg gtgcaggtct 20220ggggtgaggt caccaggccc tgcggtgagc tggttgtgcg gtgtccggtt gctgcaggtc 20280cggggtgagt tcgccaggcc ctcggtgagc tggatgtgcg gtgtccccgt gtccggatgg 20340tgcaggtcca gggtgaggtc gctaggccct tggtgggctg gatgtgccgt gtccggatgg 20400tgcaggtctg gggtgaggtc gccaggcctt tggtgagctg gatgtgcggt gtctgcatgg 20460tgcaggtctg gggtgaggtc gccaggccct tggtgggctg gatgtgtggt gtccggatgg 20520tgcaggtccg gcgtgaggtc gccaggccct gctgtgagct ggatgtgcgg tgtctggatg 20580gtgcaggtcc ggggtgaggt agccaaggcc ttcggtgagc tggatgtggg gtgtccggat 20640ggtgcaggtc cggggtgagg tcgccaggcc ctgcggttag ctggatatgc ggtgtccgga 20700tggtgcaggt ccggggtgag gtcaccaggc cctgcggtta gctggatgtg cggtgtctgg 20760atggtgcagg tccggggtga ggtcgccagg ccctgctgtg agctggatgt gctgtatccg 20820gatggtgcag gtccggggtg aggtcgccag gccctgcagt gagctggatg tgctgtatcc 20880ggatggtgca ggtctggcgt gaggtcgcca ggccctgcgg ttagctggat atgcggtgtc 20940ggatggtgca ggtccggggt gaggtcacca ggccctgcgg ttagctggat gtgcggtgtc 21000cggatggtgc aggtctgggg tgaggtcgcc aggccctgct gtgagctgga tgtgctgtat 21060ccggatggtg caggtccggg gtgaggtcgc caggccctgc ggtgagctgg atgtgctgta 21120tccggatggt gcaggtctgg cgtgaggtcg ccaggccctg cggtgagctg gatgtgcagt 21180gtacggatgg tgcaggtccg gggtgaggtc gccaggccct gcggtgggct gtatgtgtgt 21240tgtctggatg gtgcaggtcc ggggtgagtt cgccaggccc tgcggtgagc tggatgtgtg 21300gtgtctggat gctgcaggtc cggggtgagt tcgccaggcc ctcggtgagc tggatatgcg 21360gtgtccccgt gtccgaatgg tgcaggtcca gggtgaggtc gccaggccct tggtgggctg 21420gatgtgccgt gtccggatgg tgcaggtctg gggtgaggtc gccaggccct tggtgagctg 21480gatgtgcggt gtccggatgg tgcaggtccg gggtgaggtc accaggccct cggtgatctg 21540gatgtggcat gtccttctcg tttaaggggt tggctgtgtt ccggccgcag agcaccgtct 21600gcgtgaggag atcctggcca agttcctgca ctggctgatg agtgtgtacg tcgtcgagct 21660gctcaggtct ttcttttatg tcacggagac cacgtttcaa aagaacaggc tctttttcta 21720ccggaagagt gtctggagca agttgcaaag cattggaatc aggtactgta tccccacgcc 21780aggcctctgc ttctcgaagt cctggaacac cagcccggcc tcagcatgcg cctgtctcca 21840cttgcctgtg cttccctggc tgtgcagctc tgggctggga gccaggggcc ccgtcacagg 21900cctggtccaa gtggattctg tgcaaggctc tgactgcctg gagctcacgt tctcttactt 21960gtaaaatcag gagtttgtgc caagtggtct ctagggtttg taaagcagaa gggatttaaa 22020ttagatggaa acactaccac tagcctcctt gcctttccct gggatgtggg tctgattctc 22080tctctctttt ttttttcttt tttgagatgg agtctcactc tgttgcccag gctggagtgc 22140agtggcataa tcttggctca ctgcaacctc cacctcctgg gtttaagcga ttcaccagcc 22200tcagcctcct aagtagctgg gattacaggc acctgccacc acgcctggct aatttttgta 22260cttttaggag agacggggtt tcaccatgtt ggccaggctg gtctcgaact catgacctca 22320ggtgatccac ccaccttggc ctcccaaagt gctgggttta caggctaagc caccgtgccc 22380agcccccgat tctcttttaa ttcatgctgt tctgtatgaa tcttcaatct attggattta 22440ggtcatgaga ggataaaatc ccacccactt ggcgactcac tgcagggagc acctgtgcag 22500ggagcacctg gggataggag agttccacca tgagctaact tctaggtggc tgcatttgaa 22560tggctgtgag attttgtctg caatgttcgg ctgatgagag tgtgagattg tgacagattc 22620aagctggatt tgcatcagtg agggacggga gcgctggtct gggagatgcc agcctggctg 22680agcccaggcc atggtattag cttctccgtg tcccgcccag gctgactgtg gagggcttta 22740gtcagaagat cagggcttcc ccagctcccc tgcacactcg agtccctggg gggccttgtg 22800acaccccatg ccccaaatca ggatgtctgc agagggagct ggcagcagac ctcgtcagag 22860gtaacacagc ctctgggctg gggaccccga cgtggtgctg gggccatttc cttgcatctg 22920ggggagggtc agggctttcc ctgtgggaac aagttaatac acaatgcacc ttacttagac 22980tttacacgta tttaatggtg tgcgacccaa catggtcatt tgaccagtat tttggaaaga 23040atttaattgg ggtgaccgga aggagcagac agacgtggtg gtccccaaga tgctccttgt 23100cactactggg actgttgttc tgcctggggg gccttggagg cccctcctcc ctggacaggg 23160taccgtgcct tttctactct gctgggcctg cggcctgcgg tcagggcacc agctccggag 23220cacccgcggc cccagtgtcc acggagtgcc aggctgtcag ccacagatgc ccaggtccag 23280gtgtggccgc tccagccccc gtgcccccat gggtggtttt gggggaaaag gccaagggca 23340gaggtgtcag gagactggtg ggctcatgag agctgattct gctccttggc tgagctgccc 23400tgagcagcct ctcccgccct ctccatctga agggatgtgg ctctttctac ctgggggtcc 23460tgcctggggc cagccttggg ctaccccagt ggctgtacca gagggacagg catcctgtgt 23520ggaggggcat gggttcacgt ggccccagat gcagcctggg accaggctcc ctggtgctga 23580tggtgggaca gtcaccctgg gggttgaccg ccggactggg cgtccccagg gttgactata 23640ggaccaggtg tccaggtgcc ctgcaagtag aggggctctc agaggcgtct ggctggcatg 23700ggtggacgtg gccccgggca tggccttcag cgtgtgctgc cgtgggtgcc ctgagccctc 23760actgagtcgg tgggggcttg tggcttcccg tgagcttccc cctagtctgt tgtctggctg 23820agcaagcctc ctgaggggct ctctattgca gacagcactt gaagagggtg cagctgcggg 23880agctgtcgga agcagaggtc aggcagcatc gggaagccag gcccgccctg ctgacgtcca 23940gactccgctt catccccaag cctgacgggc tgcggccgat tgtgaacatg gactacgtcg 24000tgggagccag aacgttccgc agagaaaaga gggtggctgt gctttggttt aacttccttt 24060ttaaacagaa gtgcgtttga gccccacatt tggtatcagc ttagatgaag ggcccggagg 24120aggggccacg ggacacagcc agggccatgg cacggcgcca acccatttgt gcgcacagtg 24180aggtggccga ggtgccggtg cctccagaaa agcagcgtgg gggtgtaggg ggagctcctg 24240gggcagggac aggctctgag gaccacaaga agcagccggg ccagggcctg gatgcagcac 24300ggcccgaggt cctggatccg tgtcctgctg tggtgcgcag cctccgtgcg cttccgctta 24360cggggcccgg ggaccaggcc acgactgcca ggagcccacc gggctctgag gatcctggac 24420cttgccccac ggctcctgca ccccacccct gtggctgcgg tggctgcggt gaccccgtca 24480tctgaggaga gtgtggggtg aggtggacag aggtgtggca tgaggatccc gtgtgcaaca 24540cacatgcggc caggaacccg tttcaaacag ggtctgagga agctgggagg ggttctaggt 24600cccgggtctg ggtggctggg gacactgggg aggggctgct tctcccctgg gtccctatgg

24660tggggtgggc acttggccgg atccactttc ctgactgtct cccatgctgt ccccgccagg 24720ccgagcgtct cacctcgagg gtgaaggcac tgttcagcgt gctcaactac gagcgggcgc 24780ggcgccccgg cctcctgggc gcctctgtgc tgggcctgga cgatatccac agggcctggc 24840gcaccttcgt gctgcgtgtg cgggcccagg acccgccgcc tgagctgtac tttgtcaagg 24900tgggtgccgg ggacccccgt gagcagccct gctggacctt gggagtggct gcctgattgg 24960cacctcatgt tgggtggagg aggtactcct gggtgggccg cagggagtgc aggtgaccct 25020gtcactgttg aggacacacc tggcacctag ggtggaggcc ttcagccttt cctgcagcac 25080atggggccga ctgtgcaccc tgactgcccg ggctcctatt cccaaggagg gtcccactgg 25140attccagttt ccgtcagaga aggaaccgca acggctcagc caccaggccc cggtgccttg 25200caccccagtc ctgagccagg ggtctcctgt cctgaggctc agagagggga cacagcccgc 25260cctgcccttg gggtctggag tggtgggggt cagagagaga gtgggggaca ccgccaggcc 25320aggccctgag ggcagaggtg atgtctgagt ttctgcgtgg ccactgtcag tctcctcgcc 25380tccactcaca caggtggatg tgacgggcgc gtacgacacc atcccccagg acaggctcac 25440ggaggtcatc gccagcatca tcaaacccca gaacacgtac tgcgtgcgtc ggtatgccgt 25500ggtccagaag gccgcccatg ggcacgtccg caaggccttc aagagccacg taaggttcac 25560gtgtgatagt cgtgtccagg atgtgtgtct ctgggatatg aatgtgtcta gaatgcagtc 25620gtgtctgtga tgcgtttctg tggtggaggt acttccatga tttacacatc tgtgatatgc 25680gtgtgtggca cgtgtgtgtc gtggtgcatg tatctgtggc gtgcatattt gtggtgtgtg 25740tgtgtgtggc acgtgtgtgt ccatggtgtg tgtgcctgtg gtgtgcatgt gtgtgtgtct 25800gtgacacgtg catgttcatg ctgtgtgctg catgtctgtg atgtgcctat ttgtggtgtg 25860tgtgtgcatg tgtccgtgac atatgcgtgt ctatggcatg ggtgtgtgtg gccccttggc 25920cttactcctt cctcctccag gcatggtccg caccattgtc ctcacgctct cgggtgctgg 25980tttggggagc tccacattca gggtcctcac ttctagcatg ggtgcccctg tcctgtcaca 26040gggctgggcc ttggagactg taagccaggt ttgagaggag agtagggatg ctggtggtac 26100cttcctggac ccctggcacc cccaggaccc cagtctggcc tatgccggct ccatgagata 26160taggaaggct gattcaggcc tcgctccccg ggacacactc ctcccagagc ggccgggggc 26220cttggggctc ggcaggggtg aaaggggccc tgggcttggg ttcccaccca gtggtcatga 26280gcacgctgga ggggtaagcc ctcaaagtcg tgccaggccg gggtgcagag gtgaagaagt 26340atccctggag cttcggtctg gggagaggca catgtggaaa cccacaagga cctctttctc 26400tgacttcttg agcttgtggg attggttttc atgtgtggga taggtgggga tctgtgggat 26460tggtttttat gagtggggta acacagagtt caaggcgagc tttcttcctg tagtgggtct 26520gcaggtgctc caacagcttt attgaggaga ccatatcttc ctttgaacta tggtcgggtt 26580tatagtaagt caggggtgtg gaggcctccc ctgggctccc tgttctgttt cttccactct 26640ggggtcgtgt ggtgcctgct gtggtgtgtg gccggtgggc agggcttcca ggcctccttg 26700tgttcattgg cctggatgtg gccctggcta cgctccgtcc ttggaattcc cctgcgagtt 26760ggaggctttc tttctttctt tttttctttc tttttttttt tttttgataa cagagtctcg 26820ctcttttttg cccaggctgg agtggtttgg cgtgatcttg gctcactgca acctgtgctt 26880cctgagttca agcaattctc ttgcctcagc ctcccaagta gctggaatta taggcgccca 26940ccaccatgct gactaatttt tgtaatttta gtagagacga ggtttctcca tgttggccag 27000gctggtctcg aactcctgac ctcaggtgat cctcccacct cggcctccca aagtgctggg 27060atgacaggtg tgaaccgccg cgcccggccg agactcgctt cctgcagctt ccgtgagatc 27120tgcagcgata gctgcctgca gccttggtgc tgacaacctc cgttttcctt ctccaggtct 27180cgctaggggt ctttccattt catgactctc ttcacagaag agtttcacgt gtgctgattt 27240cccggctgtt tcctgcgtaa ttggtgtctg ctgtttatcg atggcctcct tccatttcct 27300ttaggctttg tttattgttg tttttccggc tccttgaagg aaaagtttcg attatggatg 27360tttgaacttt cttttctaaa caagcatctg aagttgccgt tttccctcta aagcagggat 27420cccgaggccc ctggctgtgg agtggcaccg gtctggggcc tgttaggaac ccggcgcaca 27480gcgggaggct aggtggggtg tggggagcca gcgttcccgc ctgagccccg cccctctcag 27540atcagcagtg gcatgcggtg ctcagaggcg cacacaccct actgagaact gtgcgtgaga 27600ggggtctaga ttctgtgctc cttatgggaa tctaatgcct gatgatctga ggtggaaccg 27660tttgctccca aaaccatccc cttccccact gctgtcctgt ggaaaaatcg tcttccacga 27720aaccagtccc tggtaccaca atggttgggg accctgtgct aaagacctgc ttcagcagcc 27780tctcgtcagt gttgatatat tggcttttct gtgttgagtc cagaataatt acggatttct 27840gtgatgcttt ccgccgacct cagacccatg ggctatttgt gggcgtgttg cctgctcctg 27900ggttgggaag ggtgcaggcc ccatgtacct tcctgttact gccttccagg ttggttctca 27960gggttgaatc gtactcgatg tggttttagc ccacggccct gccgccagct cctgggggct 28020ggggaacatg ctgaagcaca gagtcaccgt gcgcgtcttt tgatgcctca caagctcgag 28080gcctcctgtg tccgtgttag tgtgtgtcac gtgcctgctc acatcctgtc ttggggacgc 28140aggggcttag caggtcccgt agtaaatgac aagcgtcctg ggggagtctg cagaatagga 28200ggtgggggtg ccggtctctc tcccgcgtct tcagactctt ctcctgcctg tgctgtggct 28260gcacctgcat ccctgcaatc cctccagcac tgggctggag aggcccggga gctcgagtgc 28320cacttgtgcc acgtgactgt ggatggcagt cggtcacggg ggtctgatgt gtggtgactg 28380tggatggcgg ttggtcacag gggtctgatg tgtggtgact gtggatggcg gtcgtggggt 28440ctgatgtggt gactgtggat ggcggtcgtg gggtctgatg tgtggtgact gtggatggcg 28500gtcgtggggt ctgatgtggt gactgtggat ggcggtcgtg gggtctgatg tggtgactgt 28560ggatggcggt cgtggggtct gatgtggtga ctgtggatgg cagtcgtggg gtctgatgtg 28620tggtgactgt ggatggcggt cgtggggtct gatgtggtga ctgtggatgg cagtcgtggg 28680gtctgatgtg tggtgactgt ggatggcggt cgtggggtct gatgtgtggt gactgtggat 28740ggcggtcgtg gggtctgatg tgtggtgact gtggatggcg gtcgtggggt ctgatgtgtg 28800gtgactgtgg atggcggtcg tggggtctga tgtggtgact gtggatggcg gtcgtggggt 28860ctgatgtgtg gtgactgtgg atggtgatcg gtcacagggg tctgatgtgt ggtgactgtg 28920gatggcggtc gtggggtctg atgtgtggtg actgtggatg gtgatcggtc acaggggtct 28980gatgtgtggt gactgtggat ggcggtcgtg gggtctgatg tgtggtgact gtggatggcg 29040gttggtcccg ggggtctgat gtgtggtgac tgtggatggc gatcggtcac aggggtctga 29100tgtgtggtga ctgtggatgg cggtcgtggg gtctgatgtg tggtgactgt ggatggcggt 29160cgtggggtct gatgtgtggt gactgtggat ggcggtcgtg gggtctgatg tggtgactgt 29220ggatggcggt cgtggggtct gatgtggtga ctgtggatgg cggtcgtggg gtctgatgtg 29280tggtgactgt ggatggcggt tggtcccggg ggtctgatgt gtggtgactg tggatggcgg 29340tcgtggggtc tgatgtggtg actgtggatg gcagtcgtgg ggtctgatgt gtggtgactg 29400tggatggcgg tcgtggggtc tgatgtgtgg tgactgtgga tggcggtcgt ggggtctgat 29460gtgtggtgac tgtggatggc ggtcgtgggg tctgatgtgt ggtgactgtg gatggcggtc 29520gtggggtctg atgtggtgac tgtggatggc ggtcgtgggg tctgatgtgt ggtgactgtg 29580gatggtgatc ggtcacaggg gtctgatgtg tggtgactgt ggatggcggt cgtggggtct 29640gatgtgtggt gactgtggat ggcggtcgtg gggtctgatg tggtgactgt ggatggcggt 29700cgtggggtct gatgtgtggt gactgtggat ggcggtcgta gggtctgatg tgtggtgact 29760gtggatggca gtcggtcaca ggggtctgat gtgtggtgac tgtggatggc ggtcgtgggg 29820tctgatgtgt ggtgactgtg gatggcggtc gtggggtctg atgtgtggtg actgtggatg 29880gcggtcgtgg ggtctgatgt gtggtgactg tggatggcgg tcgtggggtc tgatgtggtg 29940actgtggatg gtgatcggtc acaggggtct gatgtgtggt agctgcaggt ggagtcccag 30000gtgtgtctgt agctactttg cgtcctcggc cccccggccc ccgtttccca aacagaagct 30060tcccaggcgc tctctgggct tcatcccgcc atcgggcttg gccgcaggtc cacacgtcct 30120gatcggaaga aacaagtgcc cagctctggc cggggcaggc cacatttgtg gctcatgccc 30180tctcctctgc cggcaggtct ctaccttgac agacctccag ccgtacatgc gacagttcgt 30240ggctcacctg caggagacca gcccgctgag ggatgccgtc gtcatcgagc aggtctgggc 30300actgccctgc agggttgggc acggactccc agcagtgggt cctcccctgg gcaatcactg 30360ggctcatgac cggacagact gttggccctg gggggcagtg gggggaatga gctgtgatgg 30420gggcatgatg agctgtgtgc cttggcgaaa tctgagctgg gccatgccag gctgcgacag 30480ctgctgcatt caggcacctg ctcacgtttg actgcgcggc ctctctccag ttccgcagtg 30540cctttgttca tgatttgcta aatgtcttct ctgccagttt tgatcttgag gccaaaggaa 30600aggtgtcccc ctcctttagg agggcaggcc atgtttgagc cgtgtcctgc ccagctggcc 30660cctcagtgct gggtctgagg ccaaaggaaa cgtgtccccc ttcttaggag gacgggccgt 30720gtttgagcca cgccccgctg agcgggcctc tcagtgctgg gtctgtccac gtggccctgt 30780ggccctttgc agatgtggtc tgtccacgtg gccctgtggc tctttgcaga tgcctgttag 30840cacttgctcg gctctagggg acagtcgtgt ccaccgcatg aggctcagag acctctgggc 30900gaatttcctt ggctcccagg gtgggggtgg aggtggcctg ggctgctggg acccagaccc 30960tgtgcccggc agctgggcag caactcctgg atcacatatg ccatccgggc cacggtgggc 31020tgtgtgggtg tgagcccagc tggacccaca ggtggcccag aggagacgtt ctgtgtcaca 31080cactctgcct aagcccatgt gtgtctgcag agactcggcc cggccagccc acgatggccc 31140tgcattccag cccagccccg cacttcatca caaacactga ccccaaaagg gacggagggt 31200cttggccacg tggtcctgcc tgtctcagca cccaccggct cactcccatg tgtctcccgt 31260ctgctttcgc agagctcctc cctgaatgag gccagcagtg gcctcttcga cgtcttccta 31320cgcttcatgt gccaccacgc cgtgcgcatc aggggcaagt gagtcaggtg gccaggtgcc 31380attgccctgc gggtggctgg gcgggctggc agggcttctg ctcacctctc tcctgcccct 31440tccccactgn ccttctgccc ggggccacca gagtctcctt ttctggcccc cgccccctcc 31500ggctcctggg ctgcaggctc ccgaggcccc ggaaacatgg ctcggcttgc ggcagccgga 31560gcggagcagg tgccacacga ggcctggaaa tggcaagcgg ggtgtggagt tgctcctgcg 31620tggaggacga ggggcggggg gtgtgtctgg gtcaggtgtg cgccgagcgt ttgagcctgc 31680agcttgtcag ctccaagtta ctactgacgc tggacacccg gctctcacac gcttgtatct 31740ctctctcccg atacaaaagg attttatccg attctcattc ctgtccctgt cgtgtgaccc 31800ccgcgagggc gcgggctctt ctctctgtga ctagatttcc catctggaaa gtgcggggtt 31860gaccgtgtag tttgctcctc tcggggggcc tgtggtggcc atggggcagg cggcctggga 31920gagctgccgt cacacagcca ctgggtgagc cacactcacg gtggtagagc cacagtgcct 31980ggtgccacat cacgtcctct ggattttaag taaaaccaca cacctcccgg caggcatctg 32040cctgcgaccc tgtgtgtgcc tggggagagt ggtagcacgg aggaaattcg tgcacactca 32100aggtcatcag caaggtcatc cgcagtcagg tggaacgtgg aggcctctct ctgggatcgt 32160ctccagcgga taaaggactg tgcacagctt cggaagcttt tatttaaaaa tataactatt 32220aattattgca ttataagtaa tcactaatgg tatcagcaat tataatattt attaaagtat 32280aattagaaat attaagtagt acacacgttc tggaaaaaca caaattgcac atggcagcag 32340agtgaatttt ggccgaggga cacgtgtgca catgtgtgta agcggccccc aggcccacag 32400aattcgctga caaagtcacc tccccagaga agccaccacg ggcctccttc gtggtcgtga 32460attttattaa gatggatcaa gtcacgtacc gtccacgtgt ggcagggctt tggggaatgt 32520gaggtgatga ctgcgtcctc atgccctgac agacaggagg tgactgtgtc tgtcctgtcc 32580ctaggacacg gacaggcccg aagctctagt ccccatcgtg gtccagtttg gcctctgaat 32640aaaaacgtct tcaaaacctg ttgccccaaa aactaagaac agagagagtt tcccatccca 32700tgtgctcaca ggggcgtatc tgcttgcgtt gactcgctgg gctggccgga ctcctagagt 32760tggtgcgtgt gcttctgtgc aaaaagtgca gtcctcttgc ccatcactgt gatatctgca 32820ccagcaagga aagcctcttt tcttttcttt cttttttttt ttttgagacg gaacgtcact 32880gttgtctgcc tgggcttgag tgcagtggcg cgatctcaac tcactgcaac ctccgcctcc 32940cgggttccag catttctcct gcctcagcct cccgagcagc tgagattaca ggcacccacc 33000ccctgcgcct ggctaatttt tgtattttta gtagagaggg gtttttgcca tgttggccag 33060gctggtctcg aactcctgac ctcaggtgat ccacccacct cggcctccca aagtgctggg 33120attacaggtg tgagccatca cgcccagccg gaaagcctct ttttaaggtg accacctata 33180gcgcttcccg aaaataacag gtcttgtttt tgcagtaggc tgcaagcgtc tcttagcaac 33240aggagtggcg tcctgtgggc tctggggatg gctgagggtc gcgtggcagc catgccttct 33300gtgtgcacct ttaggttcca cggggctatt ctgctctcac tgtttgtctg aaaacgcacc 33360cttggcatcc ttgtttggag agtttctgct tctcgttggt catgctgaaa ctaggggcaa 33420ggttgtatcc gttggcgcgc agcggctaca tgtagggtca tgagtctttc accgtggaca 33480aattccttga aaaaaaaaaa aggagtccgg ttaagcattc attccgggtc aagtgtctgg 33540ttctgtgaat aaactctaag atttaagaaa ccttaatgaa agaaaacctt gatgattcag 33600agcaaggatg tggtcacacc tgtggctgga tctgtttcag ccgccccagt gcatggtgag 33660agtggggagc agggattgtt tgttcagagg tctcatctgg tatgtttctg aggtgtttgc 33720cggctgaatg gtagacgtgt cgtttgtgtg tatgaggttc tgtgtctgtg tgtggctcgg 33780tttgagtgta cgcatgtcca gcacatgccc tgcccgtctc tcacctgtgt cttcccgccc 33840caggtcctac gtccagtgcc aggggatccc gcagggctcc atcctctcca cgctgctctg 33900cagcctgtgc tacggcgaca tggagaacaa gctgtttgcg gggattcggc gggacgggtg 33960aggcctcctc ttccccaggg gggcttgggt gggggttgat ttgcttttga tgcattcagt 34020gttaatattc ctggtgctct ggagaccatg actgctctgt cttgaggaac cagacaaggt 34080tgcagcccct tcttggtatg aagccgcacg ggaggggttg cacagcctga ggactgcggg 34140ctccacgcag gctctgtcca gcggccatgt ccagaggcct cagggctcag caggcgggag 34200ggccgctgcc ctgcatgatg agcatgtgaa ttcaacaccg aggaagcaca ccagcttctg 34260tcacgtcacc caggttccgt tagggtcctt ggggagatgg ggctggtgca gcctgaggcc 34320ccacatctcc cagcaggccc tcgacaggtg gcctggactg ggcgcctctt cagcccattg 34380cccatcccac ttgcatgggg tctacaccca aggacgcaca cacctaaata tcgtgccaac 34440ctaatgtggt tcaactcagc tggcttttat tgacagcagt tacttttttt tttttaatac 34500tttaagttct agggtacatg tgcacgacgt gcaggttagt tacatatgta tacatgtgcc 34560atgttggtgt gctgcaccca ttaactcatc atttacatta ggtatatctc ctaatgctat 34620ccctccccac tccccccatc ccatgacagg ccctggtgtg tgatgttccc caccctgtgt 34680ccaagtgttc tcattgttca gttcccacct gtgagtgaga acatgtggtg tttggttttc 34740tttccttgca atagtttgct cagagtgatg gtttccagct tcgtccatgt ccctacaaag 34800gacatgaact catccttttt tatgactgca tagtattccg tggtgtatat gtgccacatt 34860ttcttaatcc agtctatcat cgatggacat ttgggttggt tgcaagtctt tgctactgtg 34920aatagtgccg caataaacat acgtgtgcat gtgtctttat agcagcatga tttataatcc 34980tttgggtata tacccagtaa tgggatggct gggtcaaatg gtatttctag ttctagatcc 35040ttgaggaatc accacactgt cttccacaat ggttgaacta gtttacactc ccaccaacag 35100tgtaaaagtg ttctggtgct ggagaggatg tggacagcag ttattttttt atgaaaatag 35160tatcactgaa caagcagaca gttagtgaag gatgcgtcag gaagcctgca ggccacacag 35220ccatttctct cgaagactcc gggtttttcc tgtgcatctt ttgaaactct agctccaatt 35280atagcatgta cagtggatca aggttcttct tcattaaggt tcaagttcta gattgaaata 35340agtttatgta acagaaacaa aaatttcttg tacacacaac ttgctctggg atttggagga 35400aagtgtcctc gagctggcgg cacactggtc agccctctgg gacaggatac ctctggccca 35460tggtcatggg gcgctgggct tgggcctgag ggtcacacag tgcaccatgc ccagcttcct 35520gtggatagga tctgggtctc ggatcatgct gaggaccaca gctgccatgc tggtaaaggg 35580caccacgtgg ctcagagggg gcgaggttcc cagccccagc tttcttaccg tcttcagtta 35640tttttcccta agagtctgag aagtggggcc gcgcctgatg gccttcgttc gtcttcagct 35700ggcacagaat tgcacaagct gatggtaaac actgagtact tataatgaat gaggaattgc 35760tgtagcagtt aactgtagag agctcgtctg ttggaaagaa atttaagttt ttcatttaac 35820cgctttggag aatgttactt tatttatggc tgtgtaaatt gtttgacatt cagtccctcg 35880tagacagata ctacgtaaaa agtgtaaagt taaccttgct gtgtattttc ccttatttta 35940ggctgctcct gcgtttggtg gatgatttct tgttggtgac acctcacctc acccacgcga 36000aaaccttcct caggtgaggc ccgtgccgtg tgtctgtggg gacctccaca gcctgtgggc 36060tttgcagttg agccccccgt gtcctgcccc tggcaccgca gcgttgtctc tgccaagtcc 36120tctctctctg ccggtgctgg atccgcaaga gcagaggcgc ttggccgtgc acccaggcct 36180gggggcgcag gggcaccttc gggagggagt gggtaccgtg caggccctgg tcctgcagag 36240acgcacccag gttacacacg tggtgagtgc aggcggtgac ctggctcctg ctgctctttg 36300gaaagtcaag agtggcggct cctggggccc cagtgagacc cccaggagct gtgcacaggg 36360cctgcagggc cgaggcggca gcctcctccc cagggtgcac ctgagcctgc ggagagcagg 36420agctgctgag tgagctggcc cacagcgttc gctgcggtca cgttcctgcg tggggttgtt 36480tgggatcggt gggagaattt ggatttgctg agtgctgctg tcttgaacca cggagatggc 36540taggagtggg tttcagagtt gatttttgtg aatcaaacta aaatcaggca caggggacct 36600ggcctcagca caggggattg tccaatgtgg tccccctcaa gggcgcccca cagagccggt 36660gggcttgttt taaagtgcga tttgacgagg gacgagaaac cttgaaagct gtaaagggaa 36720ccctcagaaa atgtggccgc caggggtggt ttcaggtgct ttgctgggct gtgtttgtga 36780aaacccattt ggacccgccc tccaagtcca ccctccaggt ccaccctcca gggccgccct 36840gggctggggg tatgcctggc gttccttgtg ccgcagcccg gagcacagca ggctgtgcac 36900atttaaatcc actaagattc actcgggggg agcccaggtc ccaagcaact gagggctcag 36960gagtcctgag gctgctgagg ggacagagca gacggggaac gctgcttctg tgtggcaagt 37020tcctgagggt gctggccagg gaggtggctc agagtgtatg ttggggtccc accgggggca 37080gaactctgtc tctgatgagt cggcagccat gtaacaggaa ggggtggcca cagggagctg 37140ggaatgcacc aggggagctg cgcagctggc cgaggtccca gggccaggcc acaggaaggg 37200cagggggacg cccggggcca cagcagaggc cgcaggaagg gaaggggatg cccaggccag 37260agcagaggct accgggcaca ggggggctcc ctgagctggg tgagcgaggc tcatgactcg 37320gcgagggaac ctccttgacg tgaagctgac gactggtgtt gcccagctca cagcccagcc 37380aggtcccgcg cctgagcagg aactcagaac cctccccttt gtctaaagca cagcagatgc 37440cttcagggca tctaggagaa aacaggcaaa gtcgttgaga aacgtcttaa aagaaggtgg 37500gatggtggca atttcttgtc cagattttag tctgccccgg accacagatg agtctataac 37560gggattgtgg tgttgccatg gggacacatg agatggacca tcacagaggc cactggggct 37620gcacctccca tctgagtcct ggctgtcccg ggtccaggcc aggttcttgc atgctcacct 37680acctgtcctg cccgggagac agggaaagca ccccgaagtc tggagcaggg ctgggtccag 37740gctcctcaga gctcctgcca ggcccagcac cctgctccaa atcaccactt ctctggggtt 37800ttccaaagca tttaacaagg gtgtcaggtt acctcctggg tgacggcccc gcatcctggg 37860gctgacattg cccctctgcc ttaggaccct ggtccgaggt gtccctgagt atggctgcgt 37920ggtgaacttg cggaagacag tggtgaactt ccctgtagaa gacgaggccc tgggtggcac 37980ggcttttgtt cagatgccgg cccacggcct attcccctgg tgcggcctgc tgctggatac 38040ccggaccctg gaggtgcaga gcgactactc caggtgagcg cacctggccg gaagtggagc 38100ctgtgcccgg ctggggcagg tgctgctgca gggccgttgc gtccacctct gcttccgtgt 38160ggggcaggcg actgccaatc ccaaagggtc agaggccaca gggtgcccct cgtcccatct 38220ggggctgagc agaaatgcat ctttctgtgg gagtgagggt gctcacaacg ggagcagttt 38280tctgtgctat tttggtaaaa ggaaatggtg caccagacct gggtgcactg aggtgtcttc 38340agaaagcagt ctggatccga acccaagacg cccgggccct gctgggcgtg agtctctcaa 38400acccgaacac aggggccctg ctgggcatga gtccctctga acccgagacc ctggggccct 38460gctgggcgtg agtctctccg aacccagaga cttcagggcc cttttgggcg tgagtctctc 38520cgctgtgagc cccacactcc aaggctcatc cacagtctac aggatgccat gagttcatga 38580tcacgtgtga cccatcaggg gacagggcca tggtgtgggg ggggtctcta caaaattctg 38640gggtcttgtt tccccagagc ccgagagctc aaggccccgt ctcaggctca gacacaaatg 38700aattgaagat ggacacagat gcagaaatct gtgctgtttc ttttatgaat aaaaagtatc 38760aacattccag gcagggcaag gtggctcaca cctataatcc cagcactttg ggaggccgag 38820gtgggtggat cacttgaggc caggagtttg aggccaacct aaccaacata gtgaaattcc 38880atttctactt aaaaaataca aaaattagcc tggcctggtg gcacacgcct gtagtccccg 38940ctatgcggga ggctgaggca ggagaatcat ttgaacccag gaggcagagg ttgcagtgag 39000ccgagatcac accactgcac tccagcctgg gcaacagagt gagacttcat cttaaaaaaa 39060aaaaaaaaag tatcagcatt ccaaaaccat agtggacagg tgttttttta ttctgtcctt 39120cgataatatt tactggtgct gtgctagagg ccggaactgg gggtgccttc ctctgaaagg 39180cacaccttca tgggaagaga aataagtggt gaatggttgt taaaccagag gtttaaactg 39240gggtcctgtc gttctgagtt aacagtccag atctggactt tgcctctttc cagaatgctc 39300cctggggttt gcttcatggg ggagcagcag gtgtggacac cctcgtgatg ggggagcagc 39360aggtgcagac gccctcatga tgggggagtg gcaggtgcag acacccttgt gcatggtgcc 39420cagcatgtcc ctgttgcagc tccctcccca caaggatgcc ggtctcctgt gctccccaca 39480gtccctgctt ccctctcaca gccttacctg gtcctggcct ccactggctt tgtctgcatg 39540atttccacat ttcctgggct cccagcacct cttcgcctct cccaggcacc tctgcagtgc 39600tggccatacc agtcagctgt gaactgtcca ctgcttattt tgctccccat gaaatgtatt 39660ttttaggaca ggcacccctg gttccagcct ctggcacagc atcagtgaat gttattgaag

39720gacaaaggac agacaaacaa atcaggaaaa tgggttctct ctaaacacat tgcaaagcca 39780cagaggctag tgcaggatgg gtgggcatca ggtcatcaga tgtgggtcca atgccagaat 39840attctgtgct cccaaaggcc acttggtcag agtgtgtgct tgcagaggtg gctctaaaag 39900ctcagcagtg gaggcagtgg ttcgccatac tcagggtgaa ctcacatcct ctgtgtctga 39960agtatacagc agaggcttga agggcatctg ggagaagaaa acaggcaaaa tgattaagaa 40020aagtgaaaaa ggaaaagtgg taagatggga attttcttgt ccagatttta gtctcccaaa 40080ccacagctca gatggtagaa tgtggtcaga actgatggac agaacaatag aacaaaacgg 40140aagccctatc tctcagaaac gtgtgttaat gtggtatgtg gcacagctga tggaaaagag 40200agtgtgtgtg taattttttt ttctgagaaa actgactgga agcaaataag ttgtgtcttt 40260acagcatata ccagagcaga ttctaggtag aagaggagac acatgcaaac aacaccagca 40320acagaaataa aacaaaagac tcaaagggaa gggaggtgaa cgttccctgg tttggtgttg 40380gggaaggaca cacagggagg cggatgaaac cagtgaggca acgggcattg ctttcactgc 40440agagaaactc agcttgcctg agccacagtg aaaatggcca ttccctggag cgtttgtgca 40500cgtgatttat ttaaggcgcc ctgtgaggtc ctgcacattc atcctctcac tttgttctcc 40560taaccacctg agaggtagag gaggaaaggc tccaggggag cagccgccct tggtcaccca 40620gctggcaaag ggcatgcatg attgcagcct ggcctcctgc tccggggccc ttgctctgcc 40680cgaggacccc acacaagtca gacccatagg ctcagggtga gccggagccc aaggtcgtgt 40740tggggatggc tgtgaaagaa gaaatggacg tctgatgcac acttgggaag gtcctaccag 40800cagcgtcaaa gaaatgcatg tgaaactgac agcgagaccc atccctcaaa gaaacgcacg 40860tgaaactgat ggcgagacct gtccccatcc ctcatgctgg ctccttttct gggcttgcca 40920agagccagca tcaggttgag gcaagctgga aagacttttc tggaaagcag cttgtttgca 40980tggaagtcct cacaatgtcc tgtgtcttcc cagtaattcc acttctgaag tgaccagaca 41040ttatcacggg tcttatttac catttccagt gttccaggca gggggacttg ccacagcaag 41100tcacgaacct gcccaaatac agggctaagg agatattatg catcacaaaa cttgctctgc 41160cattaaacat ttttcaaaga atttttgaag aatgtttaat ggcacaaaac gtttatttca 41220atgtagcagt gttcaaagct ggatgtaaaa gaacacaccc caggagcctg ccgtgaatgt 41280catgtgtgtt catctttgga catggacata catgggcagt gagtggtggt gaggccctgg 41340aggacatcgg tgggatgcct ccatcctgcc cctctggaga caccatgtgt gccacgtgca 41400ctcactggag ccctgtttag ctggtgccac ctggctcttc catccctgag attcaaacac 41460agtgagattc cccacgccca actcagtgtt ctcccacaaa aaacctgagt cacacctgtg 41520ttcactcgag ggacgcccgg gagccagggc tccacagttt attatgtgtt tttggctgag 41580ttatgtgcag atctcatcag ggcagatgat gagtgcacaa acacggccgt gcgaggtttg 41640gatacactca acatcactag ccaggtcctg gtggagtttg gtcatgcaga gtctggatgg 41700catgtagcat ttggagtcca tggagtgagc acccagcccc ctcgggctgc agcgcatgcc 41760ccaggcagga caaggaagcg ggaggaaggc aggaggctct ttggagcaag ctttgcagga 41820gggggctggg tgtggggcag gcacctgtgt ctgacattcc cccctgtgtc tcagctatgc 41880ccggacctcc atcagagcca gtctcacctt caaccgcggc ttcaaggctg ggaggaacat 41940gcgtcgcaaa ctctttgggg tcttgcggct gaagtgtcac agcctgtttc tggatttgca 42000ggtgagcagg ctgatggtca gcacagagtt cagagttcag gaggtgtgtg cgcaagtatg 42060tgtgtgtgtg tgtgcgcgcg tgcctgcaag gctgatggtg actggctgca cgtaagagtg 42120cacatgtacg catatacacg tgagcacata catgtgtgca tgtgtgtaca tgaaggcatg 42180gcagtgtgtg cacaggtgtg caagggcaca agtgtgtgca catgcgaatg cacacctgac 42240atgcatgtgt gttcgtgcac agtcgtgtgg gcattcacgt gaggtgcatg cgtgtgggtg 42300tgcagtgtga gtagcatgtg tgcacataac atgtattgag gggtcctcgt gttcaccccg 42360ctaggtcctc agcaccagtg ccactcctta caggatgaga cggggtccca ggccttggtg 42420ggctgaggct ctgaagctgc agccctgagg gcattgtccc atctgggcat ccgcgtccac 42480tccctctcct gtgggcttct gtgtccactc cccctctcct gtgggcattt acatccactc 42540cactccctct ctcctgtggg catccgcgtc cactccccct ctctgtgggc atctgcgtcc 42600acctcccctc tctgtgggca tttgcgtcca ctccctctcc tggttccttc ctgtcttggc 42660cgagcctcgg gggcaggcag atgacacaga gtcttgactc gcccagggtg gttcgcagct 42720gccgggtgag ggccaggccg gatttcactg ggaagaggga tagtttcttg tcaaaatgtt 42780cctctttctt gttccatctg aatggatgat aaagcaaaaa gtaaaaactt aaaatcccag 42840agaggtttct accgtttctc actctttctt ggcgactcta ggtgaacagc ctccagacgg 42900tgtgcaccaa catctacaag atcctcctgc tgcaggcgta caggtgagcc gccaccaagg 42960ggtgcaggcc cagcctccag ggaccctccg cgctctgctc acctctgacc cggggcttca 43020ccttggaact cctgggtttt aggggcaagg aatgtcttac gttttcagtg gtgctgctgc 43080ctgtgcacag ttctgttcgc gtggctctgt gcaaagcacc tgttctccat ctctgggtag 43140tggtaggagc cggtgtggcc ccaggtgtcc ccactgtgcc tgtgcactgg ccgtgggacg 43200tcatggaggc catcccaggg cagcaggggc atggggtaaa gagatgttta tggggagtct 43260tagcagagga ggctgggaag gtgtctgaac agtagatggg agatcagatg cccggaggat 43320ttggggtctc agcaaagagg gccgaggtgg gtgcaggtga gggtcgctgg ccccaccccc 43380gggaaggtgc agcagagctg tggctcccca cacagcccgg ccagcacctg tgctctgggc 43440atggctgtgc tcctggaacg ttccctgtcc tggctggtca gggggtgccc ctgccaagaa 43500tcgacaactt tatcacagag ggaagggcca atctgtggag gccacagggc cagcttctgc 43560ctggagtcag ggcaggtggt ggcacaagcc tcggggctgt accaaagggc agtcgggcac 43620cacaggcccg ggcctccacc tcaacaggcc tcccgagcca ctgggagctg aatgccagga 43680ggccgaagcc ctcgccccat gagggctgag aaggagtgtg agcatttgtg ttacccaggg 43740ccgaggctgc gcgaattacc gtgcacactt gatgtgaaat gaggtcgtcg tctatcgtgg 43800aaacccagca agggctcacg ggagagtttt ccattacaag gtcgtaccat gaaaatggtt 43860tttaacccga gtgcttgcgc cttcatgctc tggcagggag ggcagagcca cagctgcatg 43920ttaccgcctt tgcaccagct ccagaggctt gggaccaggc tgtctcagtt ccagggtgcg 43980tccggctcag accgccctcc tctctgcctt ctctctctgc ctcaaatctt ccctcgtttg 44040catctccctg acgcgtgcct gggccctcgt gcaagctgct tgactccttt ccggaaaccc 44100ttggggtgtg ctggatacag gtgccactga ggactggagg tgtctgacac tgtggttgac 44160cccagggtcc agctggcgtg cttggggcct ccttgggcca tgatgaggtc agaggagttt 44220tcccaggtga aaactcctgg gaaactccca gggccatgtg acctgccacc tgctcctccc 44280atattcagct cagtcttgtc ctcatttccc caccagggtc tctagctccg aggagctccc 44340gtagagggcc tgggctcagg gcagggcggc tgagtttccc cacccatgtg gggacccttg 44400ggtagtcgct tgattgggta gccctgagga ggccgagatg cgatgggcca cgggccgttt 44460ccaaacacag agtcaggcac gtggaaggcc caggaatccc cttccctcga ggcaggagtg 44520ggagaacgga gagctgggcc ccgatttcac ggcagccagg ctgcagtggg cgaggctgtg 44580gtggtccacg tggcgctggg ggcggggtct gattcaaatc cgctggggct cggccttcct 44640ggcccgtgct ggccgcgcct ccacacgggc ttggggtgga cgccccgacc tctagcaggt 44700ggctatttct ccctttggaa gagagcccct cacccatgct aggtgtttcc ctcctgggtc 44760aggagcgtgg ccgtgtggca accccgggac cttaggctta tttatttgtt taaaaacatt 44820ctgggcctgg cttccgttgt tgctaaatgg ggaaaagaca tcccacctca gcagagttac 44880tgagaggctg aaaccggggt gctggcttga ctggtgtgat ctcaggtcat tccagaagtg 44940gctcaggaag tcagtgagac caggtacatg gggggctcag gcagtgggtg agatgaggta 45000cacggggggc tcaggcagtg ggtgaggcca ggtacatggg gggctcaggc actgggtgag 45060atgaggtaca cggggggctc aggcagaggg tcagaccagg tacacggggg ctctgatcac 45120acgcacatat gagcacatgt gcacatgtgc tgtttcatgg tagccaggtc tgtgcacacc 45180tgccccaaag tcccaggaag ctgagaggcc aaagatggag gctgacaggg ctggcgcggt 45240ggctcacacc tgtagtccca gcactttggg aggccgaggc gagaggatcc cttgagccca 45300ggagtttaag accagcctga gcaacatagt agaaccccat ctctatgaaa aataaaaaca 45360aaaattagct gaacatggtg gtgtgcgcct gtagttccaa tacttgggag gctgaagtgg 45420gaggatcact tgagcccagg aggtggaagc tgcagtgagc tgagattgca ccactgtact 45480gcagcctggg tgacagagtg agagcccatc tcaacaacaa caaagaagac tgacaaatgc 45540agtttcttgg aaagaaacat ttagtaggaa cttaacctac acacagaagc caagtcggtg 45600tctcggtgtc agtgagatga gatgatgggt cctcacacca tcaccccaga cccagggttt 45660atgcaccaca ggggcgggtg gctcagaagg gatgcgcagg acgttgatat acgatgacat 45720caaggttgtc tgacgaaggg caggattcat gataagtacc tgctggtaca caaggaacaa 45780tggataaact ggaaacctta gaggccttcc cggaacaggg gctaatcaga agccagcatg 45840gggggctggc atccaggatg gagctgcttc agcctccaca tgcgtgttca tacagatggt 45900gcacagaaac gcagtgtacc tgtgcacaca cagacacgca gctactcgca cacacaagca 45960cacacacaga catgcatgca tgcatccgtg tgtgtgcacc tgtgcccatg aggaaaccca 46020tgcatgtgca ttcatgcacg cacacaggca ccggtgggcc catgcccaca cccacgagca 46080ccgtctgatt aggaggcctt tcctctgacg ctgtccgcca tcctctcagg tttcacgcat 46140gtgtgctgca gctcccattt catcagcaag tttggaagaa ccccacattt ttcctgcgcg 46200tcatctctga cacggcctcc ctctgctact ccatcctgaa agccaagaac gcaggtatgt 46260gcaggtgcct ggcctcagtg gcagcagtgc ctgcctgctg gtgttagtgt gtcaggagac 46320tgagtgaatc tgggcttagg aagttcttac cccttttcgc atcaggaagt ggtttaaccc 46380aaccactgtc aggctcgtct gcccgccctc tcgtggggtg agcagagcac ctgatggaag 46440ggacaggagc tgtctgggag ctgccatcct tcccaccttg ctctgcctgg ggaagcgctg 46500gggggcctgg tctctcctgt ttgccccatg gtgggatttg gggggcctgg cctctcctgt 46560ttgccctgtg gtgggattgg gctgtctccc gtccatggca cttagggccc ttgtgcaaac 46620ccaggccaag ggcttaggag gaggccaggc ccaggctacc ccacccctct caggagcaga 46680ggccgcgtat caccacgaca gagccccgcg ccgtcctctg cttcccagtc accgtcctct 46740gcccctggac actttgtcca gcatcaggga ggtttctgat ccgtctgaaa ttcaagccat 46800gtcgaacctg cggtcctgag cttaacagct tctactttct gttctttctg tgttgtggaa 46860atttcacctg gagaagccga agaaaacatt tctgtcgtga ctcctgcggt gcttgggtcg 46920ggacagccag agatggagcc accccgcaga ccgtcgggtg tgggcagctt tccggtgtct 46980cctgggaggg gagctgggct gggcctgtga ctcctcagcc tctgttttcc cccagggatg 47040tcgctggggg ccaagggcgc cgccggccct ctgccctccg aggccgtgca gtggctgtgc 47100caccaagcat tcctgctcaa gctgactcga caccgtgtca cctacgtgcc actcctgggg 47160tcactcagga caggcaagtg tgggtggagg ccagtgcggg ccccacctgc ccaggggtca 47220tccttgaacg ccctgtgtgg ggcgagcagc ctcagatgct gctgaagtgc agacgccccc 47280gggcctgacc ctgggggcct ggagccacgc tggcagccct atgtgattaa acgctggtgt 47340ccccaggcca cggagcctgg cagggtcccc aacttcttga acccctgctt cccatctcag 47400gggcgatggc tccccacgct tgggagcctt ctgacccctg acctgtgtcc tctcacagcc 47460tcttccctgg ctgctgccct gagctcctgg ggtcctgagc aagttctctc cccgccccgc 47520cgctccagcg tcactgggct gcctgtctgc tcgccccggt ggaggggtgt ctgtcccttc 47580actgaggttc ccaccagcca gggccacgag gtgcaggccc tgcctgcccg gccacccaca 47640cgtcctagga gggttggagg atgccacctc tggcctcttc tggaacggag tctgattttg 47700gccccgcagc ccagacgcag ctgagtcgga agctcccggg gacgacgctg actgccctgg 47760aggccgcagc caacccggca ctgccctcag acttcaagac catcctggac tgatggccac 47820ccgcccacag ccaggccgag agcagacacc agcagccctg tcacgccggg ctctacgtcc 47880cagggaggga ggggcggccc acacccaggc ccgcaccgct gggagtctga ggcctgagtg 47940agtgtttggc cgaggcctgc atgtccggct gaaggctgag tgtccggctg aggcctgagc 48000gagtgtccag ccaagggctg agtgtccagc acacctgccg tcttcacttc cccacaggct 48060ggcgctcggc tccaccccag ggccagcttt tcctcaccag gagcccggct tccactcccc 48120acataggaat agtccatccc cagattcgcc attgttcacc cctcgccctg ccctcctttg 48180ccttccaccc ccaccatcca ggtggagacc ctgagaagga ccctgggagc tctgggaatt 48240tggagtgacc aaaggtgtgc cctgtacaca ggcgaggacc ctgcacctgg atgggggtcc 48300ctgtgggtca aattgggggg aggtgctgtg ggagtaaaat actgaatata tgagtttttc 48360agttttgaaa aaaatctcat gtttgaatcc taatgtgcac tgcatagaca ccactgtatg 48420caattacaga agcctgtgag tgaacggggt ggtggtcagt gcgggcccat ggcctggctg 48480tgcatttacg gaagtctatg agtgaatggg gttgtggtca gtgcgggccc atggcctggc 48540tgggcctggg aggtttctga tgctgtgagg caggagggga aggagggtag gggatagaca 48600gtgggagccc ccaccctgga agacataaca gtaagtccag gcccgaaggg cagcagggat 48660gctgggggcc cagcttgggc ggcggggatg atggagggcc tggccagggt ggcagggatg 48720atgggggccc cagctggggt ggcaggggtg atgggggggg ctggtctggg tggcggggaa 48780gatggggaag cctggctggg ccccctcctc ccctgcctcc cacctgcagc cgtggatccg 48840gatgtgcttc cctggtgcac atcctctggg ccatcagctt tcatggaggt ggggggcagg 48900ggcatgacac catcctgtat aaaatccagg attcctcctc ctgaacgccc caactcaggt 48960tgaaagtcac attccgcctc tggccattct cttaagagta gaccaggatt ctgatctctg 49020aagggtgggt agggtggggc agtggagggt gtggacacag gaggcttcag ggtggggctg 49080gtgatgctct ctcatcctct tatcatctcc cagtctcatc tctcatcctc ttatcatctc 49140ccagtctcat ctgtcttcct cttatctccc agtctcatct gtcatcctct taccatctcc 49200cagtctcatc tcttatcctc ttatctccta gtctcatcca gacttacctc ccagggcggg 49260tgccaggctc gcagtggagc tggacatacg tccttcctca ggcagaagga actggaagga 49320ttgcagagaa caggaggggc ggctcagagg gacgcagtct tggggtgaag aaacagcccc 49380tcctcagaag ttggcttggg ccacacgaaa ccgagggccc tgcgtgagtg gctccagagc 49440cttccagcag gtccctggtg gggccttatg gtatggccgg gtcctactga gtgcaccttg 49500gacagggctt ctggtttgag tgcagcccgg acgtgcctgg tgtcggggtg ggggcttatg 49560gccactggat atggcgtcat ttattgctgc tgcttcagag aatgtctgag tgaccgagcc 49620taatgtgtat ggtgggccca agtccacaga ctgtgtcgta aatgcactct ggtgcctgga 49680gcccccgtat aggagctgtg aggaaggagg ggctcttggc agccggcctg ggggcgcctt 49740tgccctgcaa actggaaggg agcggccccg ggcgccgtgg gcggacgacc tcaagtgaga 49800ggttggacag aacagggcgg ggacttccca ggagcagagg ccgctgctca ggcacacctg 49860ggtttgaatc acagaccaac aggtcaggcc attgttcagc tatccatctt ctacaaagct 49920ccagattcct gtttctccgg gtgttttttg ttgaaatttt actcaggatt acttatattt 49980tttgctaaag tattagaccc ttaaaaaagg tatttgcttt gatatggctt aactcactaa 50040gcacctactt tatttgtctg tttttattta ttattattat tattattaga gatggtgtct 50100actctgtcac ccaggttgtt agtgcagtgg cacagtcatg gctcgctgta gccgcaaacc 50160cccaggctca agtgatcctc cggcctcagc ttcccagagt gctgggatta caggtgtgag 50220ccactgccct tgcctggcac ttttaaaaac cactatgtaa ggtcaggtcc agtggcttcc 50280acacctgtca tcccagtagt ttgggaagcc gaggcagaag gattgtctga ggccaggagt 50340ttgagaccag catgggtaac atagggagac cccatctcta caaaaaatgc aaaaagttat 50400ccgggcgtgg ggtccagcat ctgtagtccc agctgctcgg gaggctgagt gggaggatcg 50460cttgagcccg ggaggtcatg gctgcagtga gctgtgattg taccatcgca ctccagcctg 50520ggcaacagag tgagaccctg tctcaaaaaa aaaaaaaaaa aaagaaggag aaggagaaga 50580gaagaagaag gaagaaggaa agagaagaag aaggaagaag gaagaaagaa ggagaaggag 50640gcctgctagg tgctaggtag actgtcaaat ctcagagcaa aatgaaaata acaaagtttt 50700aaagggaaag aaaaacccca gctctttgga cttccttagg cctgaacttc atctcaagca 50760gcttccttcc acagacaagc gtgtatggag cgagtgagtt caaagcagaa agggaggaga 50820agcaggcaag ggtggaggct gtgggtgaca ccagccagga cccctgaaag ggagtggttg 50880ttttcctgcc tcagccccac gctcctgccg gtcctgcacc tgctgtaacc gtcgatgttg 50940gtgccaggtg cccacctggg aaggatgctg tgcagggggc ttgccaaact ttggtgggtt 51000tcagaagccc caggcacttg tggcaggcac aattacagcc cctccccaaa gatgcccacg 51060tccttctcct ggaacctgtg aatgtgtcac ccgcaaggca gaggctggtg aaggctgcag 51120gtggaatcac ggctgccagt cagccgatct taaggtcatc ctggattatc tggtgggcct 51180gatatggcca caagggtccc tagaagtgag agagggaggc aggggagagt cagagagggg 51240acgtgagaag gaccactggc cactgctggc tttgagatgg aggagggggt ccccagccaa 51300ggaatggggg cagccgctcc atgctggaaa agcaagcaat cctccccggt cctgagggca 51360cacggccctg cccacgcctc gatttcaggc cagtgggacc tgtttcagct ttccggcctc 51420cagagctgta agatgatgcg tttgtgttca gccactaagc tgcagtgatt cgtcacagca 51480gcaaatggaa tagcagtaca gggaaatgaa tacagggaca gttctcagag tgactctcag 51540cccacccctg gg 5155241132PRTHomo sapiens 4Met Pro Arg Ala Pro Arg Cys Arg Ala Val Arg Ser Leu Leu Arg Ser 1 5 10 15 His Tyr Arg Glu Val Leu Pro Leu Ala Thr Phe Val Arg Arg Leu Gly 20 25 30 Pro Gln Gly Trp Arg Leu Val Gln Arg Gly Asp Pro Ala Ala Phe Arg 35 40 45 Ala Leu Val Ala Gln Cys Leu Val Cys Val Pro Trp Asp Ala Arg Pro 50 55 60 Pro Pro Ala Ala Pro Ser Phe Arg Gln Val Ser Cys Leu Lys Glu Leu 65 70 75 80 Val Ala Arg Val Leu Gln Arg Leu Cys Glu Arg Gly Ala Lys Asn Val 85 90 95 Leu Ala Phe Gly Phe Ala Leu Leu Asp Gly Ala Arg Gly Gly Pro Pro 100 105 110 Glu Ala Phe Thr Thr Ser Val Arg Ser Tyr Leu Pro Asn Thr Val Thr 115 120 125 Asp Ala Leu Arg Gly Ser Gly Ala Trp Gly Leu Leu Leu Arg Arg Val 130 135 140 Gly Asp Asp Val Leu Val His Leu Leu Ala Arg Cys Ala Leu Phe Val 145 150 155 160 Leu Val Ala Pro Ser Cys Ala Tyr Gln Val Cys Gly Pro Pro Leu Tyr 165 170 175 Gln Leu Gly Ala Ala Thr Gln Ala Arg Pro Pro Pro His Ala Ser Gly 180 185 190 Pro Arg Arg Arg Leu Gly Cys Glu Arg Ala Trp Asn His Ser Val Arg 195 200 205 Glu Ala Gly Val Pro Leu Gly Leu Pro Ala Pro Gly Ala Arg Arg Arg 210 215 220 Gly Gly Ser Ala Ser Arg Ser Leu Pro Leu Pro Lys Arg Pro Arg Arg 225 230 235 240 Gly Ala Ala Pro Glu Pro Glu Arg Thr Pro Val Gly Gln Gly Ser Trp 245 250 255 Ala His Pro Gly Arg Thr Arg Gly Pro Ser Asp Arg Gly Phe Cys Val 260 265 270 Val Ser Pro Ala Arg Pro Ala Glu Glu Ala Thr Ser Leu Glu Gly Ala 275 280 285 Leu Ser Gly Thr Arg His Ser His Pro Ser Val Gly Arg Gln His His 290 295 300 Ala Gly Pro Pro Ser Thr Ser Arg Pro Pro Arg Pro Trp Asp Thr Pro 305 310 315 320 Cys Pro Pro Val Tyr Ala Glu Thr Lys His Phe Leu Tyr Ser Ser Gly 325 330 335 Asp Lys Glu Gln Leu Arg Pro Ser Phe Leu Leu Ser Ser Leu Arg Pro 340 345 350 Ser Leu Thr Gly Ala Arg Arg Leu Val Glu Thr Ile Phe Leu Gly Ser 355 360 365 Arg Pro Trp Met Pro Gly Thr Pro Arg Arg Leu Pro Arg Leu Pro Gln 370 375 380 Arg Tyr Trp Gln Met Arg Pro Leu Phe Leu Glu Leu Leu Gly Asn His 385 390 395 400 Ala Gln Cys Pro Tyr Gly Val Leu Leu Lys Thr His Cys Pro Leu Arg 405 410 415 Ala Ala Val Thr Pro Ala Ala Gly Val Cys Ala Arg Glu Lys Pro Gln 420 425 430 Gly Ser Val Ala Ala Pro Glu Glu Glu Asp Thr Asp Pro Arg Arg Leu 435 440 445 Val Gln Leu Leu Arg Gln His Ser Ser Pro Trp Gln Val Tyr Gly Phe 450 455 460 Val Arg Ala Cys Leu Arg Arg Leu Val Pro Pro Gly Leu Trp Gly Ser 465 470 475 480 Arg His Asn Glu Arg Arg Phe Leu Arg

Asn Thr Lys Lys Phe Ile Ser 485 490 495 Leu Gly Lys His Ala Lys Leu Ser Leu Gln Glu Leu Thr Trp Lys Met 500 505 510 Ser Val Arg Asp Cys Ala Trp Leu Arg Arg Ser Pro Gly Val Gly Cys 515 520 525 Val Pro Ala Ala Glu His Arg Leu Arg Glu Glu Ile Leu Ala Lys Phe 530 535 540 Leu His Trp Leu Met Ser Val Tyr Val Val Glu Leu Leu Arg Ser Phe 545 550 555 560 Phe Tyr Val Thr Glu Thr Thr Phe Gln Lys Asn Arg Leu Phe Phe Tyr 565 570 575 Arg Lys Ser Val Trp Ser Lys Leu Gln Ser Ile Gly Ile Arg Gln His 580 585 590 Leu Lys Arg Val Gln Leu Arg Glu Leu Ser Glu Ala Glu Val Arg Gln 595 600 605 His Arg Glu Ala Arg Pro Ala Leu Leu Thr Ser Arg Leu Arg Phe Ile 610 615 620 Pro Lys Pro Asp Gly Leu Arg Pro Ile Val Asn Met Asp Tyr Val Val 625 630 635 640 Gly Ala Arg Thr Phe Arg Arg Glu Lys Arg Ala Glu Arg Leu Thr Ser 645 650 655 Arg Val Lys Ala Leu Phe Ser Val Leu Asn Tyr Glu Arg Ala Arg Arg 660 665 670 Pro Gly Leu Leu Gly Ala Ser Val Leu Gly Leu Asp Asp Ile His Arg 675 680 685 Ala Trp Arg Thr Phe Val Leu Arg Val Arg Ala Gln Asp Pro Pro Pro 690 695 700 Glu Leu Tyr Phe Val Lys Val Asp Val Thr Gly Ala Tyr Asp Thr Ile 705 710 715 720 Pro Gln Asp Arg Leu Thr Glu Val Ile Ala Ser Ile Ile Lys Pro Gln 725 730 735 Asn Thr Tyr Cys Val Arg Arg Tyr Ala Val Val Gln Lys Ala Ala His 740 745 750 Gly His Val Arg Lys Ala Phe Lys Ser His Val Ser Thr Leu Thr Asp 755 760 765 Leu Gln Pro Tyr Met Arg Gln Phe Val Ala His Leu Gln Glu Thr Ser 770 775 780 Pro Leu Arg Asp Ala Val Val Ile Glu Gln Ser Ser Ser Leu Asn Glu 785 790 795 800 Ala Ser Ser Gly Leu Phe Asp Val Phe Leu Arg Phe Met Cys His His 805 810 815 Ala Val Arg Ile Arg Gly Lys Ser Tyr Val Gln Cys Gln Gly Ile Pro 820 825 830 Gln Gly Ser Ile Leu Ser Thr Leu Leu Cys Ser Leu Cys Tyr Gly Asp 835 840 845 Met Glu Asn Lys Leu Phe Ala Gly Ile Arg Arg Asp Gly Leu Leu Leu 850 855 860 Arg Leu Val Asp Asp Phe Leu Leu Val Thr Pro His Leu Thr His Ala 865 870 875 880 Lys Thr Phe Leu Arg Thr Leu Val Arg Gly Val Pro Glu Tyr Gly Cys 885 890 895 Val Val Asn Leu Arg Lys Thr Val Val Asn Phe Pro Val Glu Asp Glu 900 905 910 Ala Leu Gly Gly Thr Ala Phe Val Gln Met Pro Ala His Gly Leu Phe 915 920 925 Pro Trp Cys Gly Leu Leu Leu Asp Thr Arg Thr Leu Glu Val Gln Ser 930 935 940 Asp Tyr Ser Ser Tyr Ala Arg Thr Ser Ile Arg Ala Ser Leu Thr Phe 945 950 955 960 Asn Arg Gly Phe Lys Ala Gly Arg Asn Met Arg Arg Lys Leu Phe Gly 965 970 975 Val Leu Arg Leu Lys Cys His Ser Leu Phe Leu Asp Leu Gln Val Asn 980 985 990 Ser Leu Gln Thr Val Cys Thr Asn Ile Tyr Lys Ile Leu Leu Leu Gln 995 1000 1005 Ala Tyr Arg Phe His Ala Cys Val Leu Gln Leu Pro Phe His Gln 1010 1015 1020 Gln Val Trp Lys Asn Pro Thr Phe Phe Leu Arg Val Ile Ser Asp 1025 1030 1035 Thr Ala Ser Leu Cys Tyr Ser Ile Leu Lys Ala Lys Asn Ala Gly 1040 1045 1050 Met Ser Leu Gly Ala Lys Gly Ala Ala Gly Pro Leu Pro Ser Glu 1055 1060 1065 Ala Val Gln Trp Leu Cys His Gln Ala Phe Leu Leu Lys Leu Thr 1070 1075 1080 Arg His Arg Val Thr Tyr Val Pro Leu Leu Gly Ser Leu Arg Thr 1085 1090 1095 Ala Gln Thr Gln Leu Ser Arg Lys Leu Pro Gly Thr Thr Leu Thr 1100 1105 1110 Ala Leu Glu Ala Ala Ala Asn Pro Ala Leu Pro Ser Asp Phe Lys 1115 1120 1125 Thr Ile Leu Asp 1130 5 6492DNAHomo sapiens 5tttctgtgaa gcagaagtct gggaatcgat ctggaaatcc tcctaatttt tactccctct 60ccccgcgact cctgattcat tgggaagttt caaatcagct ataactggag agtgctgaag 120attgatggga tcgttgcctt atgcatttgt tttggtttta caaaaaggaa acttgacaga 180ggatcatgct gtacttaaaa aatacaacat cacagaggaa gtagactgat attaacaata 240cttactaata ataacgtgcc tcatgaaata aagatccgaa aggaattgga ataaaaattt 300cctgcatctc atgccaaggg ggaaacacca gaatcaagtg ttccgcgtga ttgaagacac 360cccctcgtcc aagaatgcaa agcacatcca ataaaatagc tggattataa ctcctcttct 420ttctctgggg gccgtggggt gggagctggg gcgagaggtg ccgttggccc ccgttgcttt 480tcctctggga aggatggcgc acgctgggag aacagggtac gataaccggg agatagtgat 540gaagtacatc cattataagc tgtcgcagag gggctacgag tgggatgcgg gagatgtggg 600cgccgcgccc ccgggggccg cccccgcacc gggcatcttc tcctcccagc ccgggcacac 660gccccatcca gccgcatccc gggacccggt cgccaggacc tcgccgctgc agaccccggc 720tgcccccggc gccgccgcgg ggcctgcgct cagcccggtg ccacctgtgg tccacctgac 780cctccgccag gccggcgacg acttctcccg ccgctaccgc cgcgacttcg ccgagatgtc 840cagccagctg cacctgacgc ccttcaccgc gcggggacgc tttgccacgg tggtggagga 900gctcttcagg gacggggtga actgggggag gattgtggcc ttctttgagt tcggtggggt 960catgtgtgtg gagagcgtca accgggagat gtcgcccctg gtggacaaca tcgccctgtg 1020gatgactgag tacctgaacc ggcacctgca cacctggatc caggataacg gaggctggga 1080tgcctttgtg gaactgtacg gccccagcat gcggcctctg tttgatttct cctggctgtc 1140tctgaagact ctgctcagtt tggccctggt gggagcttgc atcaccctgg gtgcctatct 1200gggccacaag tgaagtcaac atgcctgccc caaacaaata tgcaaaaggt tcactaaagc 1260agtagaaata atatgcattg tcagtgatgt accatgaaac aaagctgcag gctgtttaag 1320aaaaaataac acacatataa acatcacaca cacagacaga cacacacaca cacaacaatt 1380aacagtcttc aggcaaaacg tcgaatcagc tatttactgc caaagggaaa tatcatttat 1440tttttacatt attaagaaaa aaagatttat ttatttaaga cagtcccatc aaaactcctg 1500tctttggaaa tccgaccact aattgccaag caccgcttcg tgtggctcca cctggatgtt 1560ctgtgcctgt aaacatagat tcgctttcca tgttgttggc cggatcacca tctgaagagc 1620agacggatgg aaaaaggacc tgatcattgg ggaagctggc tttctggctg ctggaggctg 1680gggagaaggt gttcattcac ttgcatttct ttgccctggg ggctgtgata ttaacagagg 1740gagggttcct gtggggggaa gtccatgcct ccctggcctg aagaagagac tctttgcata 1800tgactcacat gatgcatacc tggtgggagg aaaagagttg ggaacttcag atggacctag 1860tacccactga gatttccacg ccgaaggaca gcgatgggaa aaatgccctt aaatcatagg 1920aaagtatttt tttaagctac caattgtgcc gagaaaagca ttttagcaat ttatacaata 1980tcatccagta ccttaagccc tgattgtgta tattcatata ttttggatac gcacccccca 2040actcccaata ctggctctgt ctgagtaaga aacagaatcc tctggaactt gaggaagtga 2100acatttcggt gacttccgca tcaggaaggc tagagttacc cagagcatca ggccgccaca 2160agtgcctgct tttaggagac cgaagtccgc agaacctgcc tgtgtcccag cttggaggcc 2220tggtcctgga actgagccgg ggccctcact ggcctcctcc agggatgatc aacagggcag 2280tgtggtctcc gaatgtctgg aagctgatgg agctcagaat tccactgtca agaaagagca 2340gtagaggggt gtggctgggc ctgtcaccct ggggccctcc aggtaggccc gttttcacgt 2400ggagcatggg agccacgacc cttcttaaga catgtatcac tgtagaggga aggaacagag 2460gccctgggcc cttcctatca gaaggacatg gtgaaggctg ggaacgtgag gagaggcaat 2520ggccacggcc cattttggct gtagcacatg gcacgttggc tgtgtggcct tggcccacct 2580gtgagtttaa agcaaggctt taaatgactt tggagagggt cacaaatcct aaaagaagca 2640ttgaagtgag gtgtcatgga ttaattgacc cctgtctatg gaattacatg taaaacatta 2700tcttgtcact gtagtttggt tttatttgaa aacctgacaa aaaaaaagtt ccaggtgtgg 2760aatatggggg ttatctgtac atcctggggc attaaaaaaa aaatcaatgg tggggaacta 2820taaagaagta acaaaagaag tgacatcttc agcaaataaa ctaggaaatt tttttttctt 2880ccagtttaga atcagccttg aaacattgat ggaataactc tgtggcatta ttgcattata 2940taccatttat ctgtattaac tttggaatgt actctgttca atgtttaatg ctgtggttga 3000tatttcgaaa gctgctttaa aaaaatacat gcatctcagc gtttttttgt ttttaattgt 3060atttagttat ggcctataca ctatttgtga gcaaaggtga tcgttttctg tttgagattt 3120ttatctcttg attcttcaaa agcattctga gaaggtgaga taagccctga gtctcagcta 3180cctaagaaaa acctggatgt cactggccac tgaggagctt tgtttcaacc aagtcatgtg 3240catttccacg tcaacagaat tgtttattgt gacagttata tctgttgtcc ctttgacctt 3300gtttcttgaa ggtttcctcg tccctgggca attccgcatt taattcatgg tattcaggat 3360tacatgcatg tttggttaaa cccatgagat tcattcagtt aaaaatccag atggcaaatg 3420accagcagat tcaaatctat ggtggtttga cctttagaga gttgctttac gtggcctgtt 3480tcaacacaga cccacccaga gccctcctgc cctccttccg cgggggcttt ctcatggctg 3540tccttcaggg tcttcctgaa atgcagtggt gcttacgctc caccaagaaa gcaggaaacc 3600tgtggtatga agccagacct ccccggcggg cctcagggaa cagaatgatc agacctttga 3660atgattctaa tttttaagca aaatattatt ttatgaaagg tttacattgt caaagtgatg 3720aatatggaat atccaatcct gtgctgctat cctgccaaaa tcattttaat ggagtcagtt 3780tgcagtatgc tccacgtggt aagatcctcc aagctgcttt agaagtaaca atgaagaacg 3840tggacgtttt taatataaag cctgttttgt cttttgttgt tgttcaaacg ggattcacag 3900agtatttgaa aaatgtatat atattaagag gtcacggggg ctaattgctg gctggctgcc 3960ttttgctgtg gggttttgtt acctggtttt aataacagta aatgtgccca gcctcttggc 4020cccagaactg tacagtattg tggctgcact tgctctaaga gtagttgatg ttgcattttc 4080cttattgtta aaaacatgtt agaagcaatg aatgtatata aaagcctcaa ctagtcattt 4140ttttctcctc ttcttttttt tcattatatc taattatttt gcagttgggc aacagagaac 4200catccctatt ttgtattgaa gagggattca catctgcatc ttaactgctc tttatgaatg 4260aaaaaacagt cctctgtatg tactcctctt tacactggcc agggtcagag ttaaatagag 4320tatatgcact ttccaaattg gggacaaggg ctctaaaaaa agccccaaaa ggagaagaac 4380atctgagaac ctcctcggcc ctcccagtcc ctcgctgcac aaatactccg caagagaggc 4440cagaatgaca gctgacaggg tctatggcca tcgggtcgtc tccgaagatt tggcaggggc 4500agaaaactct ggcaggctta agatttggaa taaagtcaca gaattaagga agcacctcaa 4560tttagttcaa acaagacgcc aacattctct ccacagctca cttacctctc tgtgttcaga 4620tgtggccttc catttatatg tgatctttgt tttattagta aatgcttatc atctaaagat 4680gtagctctgg cccagtggga aaaattagga agtgattata aatcgagagg agttataata 4740atcaagatta aatgtaaata atcagggcaa tcccaacaca tgtctagctt tcacctccag 4800gatctattga gtgaacagaa ttgcaaatag tctctatttg taattgaact tatcctaaaa 4860caaatagttt ataaatgtga acttaaactc taattaattc caactgtact tttaaggcag 4920tggctgtttt tagactttct tatcacttat agttagtaat gtacacctac tctatcagag 4980aaaaacagga aaggctcgaa atacaagcca ttctaaggaa attagggagt cagttgaaat 5040tctattctga tcttattctg tggtgtcttt tgcagcccag acaaatgtgg ttacacactt 5100tttaagaaat acaattctac attgtcaagc ttatgaaggt tccaatcaga tctttattgt 5160tattcaattt ggatctttca gggatttttt ttttaaatta ttatgggaca aaggacattt 5220gttggagggg tgggagggag gaagaatttt taaatgtaaa acattcccaa gtttggatca 5280gggagttgga agttttcaga ataaccagaa ctaagggtat gaaggacctg tattggggtc 5340gatgtgatgc ctctgcgaag aaccttgtgt gacaaatgag aaacattttg aagtttgtgg 5400tacgaccttt agattccaga gacatcagca tggctcaaag tgcagctccg tttggcagtg 5460caatggtata aatttcaagc tggatatgtc taatgggtat ttaaacaata aatgtgcagt 5520tttaactaac aggatattta atgacaacct tctggttggt agggacatct gtttctaaat 5580gtttattatg tacaatacag aaaaaaattt tataaaatta agcaatgtga aactgaattg 5640gagagtgata atacaagtcc tttagtctta cccagtgaat cattctgttc catgtctttg 5700gacaaccatg accttggaca atcatgaaat atgcatctca ctggatgcaa agaaaatcag 5760atggagcatg aatggtactg taccggttca tctggactgc cccagaaaaa taacttcaag 5820caaacatcct atcaacaaca aggttgttct gcataccaag ctgagcacag aagatgggaa 5880cactggtgga ggatggaaag gctcgctcaa tcaagaaaat tctgagacta ttaataaata 5940agactgtagt gtagatactg agtaaatcca tgcacctaaa ccttttggaa aatctgccgt 6000gggccctcca gatagctcat ttcattaagt ttttccctcc aaggtagaat ttgcaagagt 6060gacagtggat tgcatttctt ttggggaagc tttcttttgg tggttttgtt tattatacct 6120tcttaagttt tcaaccaagg tttgcttttg ttttgagtta ctggggttat ttttgtttta 6180aataaaaata agtgtacaat aagtgttttt gtattgaaag cttttgttat caagattttc 6240atacttttac cttccatggc tctttttaag attgatactt ttaagaggtg gctgatattc 6300tgcaacactg tacacataaa aaatacggta aggatacttt acatggttaa ggtaaagtaa 6360gtctccagtt ggccaccatt agctataatg gcactttgtt tgtgttgttg gaaaaagtca 6420cattgccatt aaactttcct tgtctgtcta gttaatattg tgaagaaaaa taaagtacag 6480tgtgagatac tg 64926239PRTHomo sapiens 6Met Ala His Ala Gly Arg Thr Gly Tyr Asp Asn Arg Glu Ile Val Met 1 5 10 15 Lys Tyr Ile His Tyr Lys Leu Ser Gln Arg Gly Tyr Glu Trp Asp Ala 20 25 30 Gly Asp Val Gly Ala Ala Pro Pro Gly Ala Ala Pro Ala Pro Gly Ile 35 40 45 Phe Ser Ser Gln Pro Gly His Thr Pro His Pro Ala Ala Ser Arg Asp 50 55 60 Pro Val Ala Arg Thr Ser Pro Leu Gln Thr Pro Ala Ala Pro Gly Ala 65 70 75 80 Ala Ala Gly Pro Ala Leu Ser Pro Val Pro Pro Val Val His Leu Thr 85 90 95 Leu Arg Gln Ala Gly Asp Asp Phe Ser Arg Arg Tyr Arg Arg Asp Phe 100 105 110 Ala Glu Met Ser Ser Gln Leu His Leu Thr Pro Phe Thr Ala Arg Gly 115 120 125 Arg Phe Ala Thr Val Val Glu Glu Leu Phe Arg Asp Gly Val Asn Trp 130 135 140 Gly Arg Ile Val Ala Phe Phe Glu Phe Gly Gly Val Met Cys Val Glu 145 150 155 160 Ser Val Asn Arg Glu Met Ser Pro Leu Val Asp Asn Ile Ala Leu Trp 165 170 175 Met Thr Glu Tyr Leu Asn Arg His Leu His Thr Trp Ile Gln Asp Asn 180 185 190 Gly Gly Trp Asp Ala Phe Val Glu Leu Tyr Gly Pro Ser Met Arg Pro 195 200 205 Leu Phe Asp Phe Ser Trp Leu Ser Leu Lys Thr Leu Leu Ser Leu Ala 210 215 220 Leu Val Gly Ala Cys Ile Thr Leu Gly Ala Tyr Leu Gly His Lys 225 230 235 71207DNAHomo sapiens 7tttctgtgaa gcagaagtct gggaatcgat ctggaaatcc tcctaatttt tactccctct 60ccccgcgact cctgattcat tgggaagttt caaatcagct ataactggag agtgctgaag 120attgatggga tcgttgcctt atgcatttgt tttggtttta caaaaaggaa acttgacaga 180ggatcatgct gtacttaaaa aatacaacat cacagaggaa gtagactgat attaacaata 240cttactaata ataacgtgcc tcatgaaata aagatccgaa aggaattgga ataaaaattt 300cctgcatctc atgccaaggg ggaaacacca gaatcaagtg ttccgcgtga ttgaagacac 360cccctcgtcc aagaatgcaa agcacatcca ataaaatagc tggattataa ctcctcttct 420ttctctgggg gccgtggggt gggagctggg gcgagaggtg ccgttggccc ccgttgcttt 480tcctctggga aggatggcgc acgctgggag aacagggtac gataaccggg agatagtgat 540gaagtacatc cattataagc tgtcgcagag gggctacgag tgggatgcgg gagatgtggg 600cgccgcgccc ccgggggccg cccccgcacc gggcatcttc tcctcccagc ccgggcacac 660gccccatcca gccgcatccc gggacccggt cgccaggacc tcgccgctgc agaccccggc 720tgcccccggc gccgccgcgg ggcctgcgct cagcccggtg ccacctgtgg tccacctgac 780cctccgccag gccggcgacg acttctcccg ccgctaccgc cgcgacttcg ccgagatgtc 840cagccagctg cacctgacgc ccttcaccgc gcggggacgc tttgccacgg tggtggagga 900gctcttcagg gacggggtga actgggggag gattgtggcc ttctttgagt tcggtggggt 960catgtgtgtg gagagcgtca accgggagat gtcgcccctg gtggacaaca tcgccctgtg 1020gatgactgag tacctgaacc ggcacctgca cacctggatc caggataacg gaggctgggt 1080aggtgcactt ggtgatgtga gtctgggctg aggccacagg tccgagatgc gggggttgga 1140gtgcgggtgg gctcctgggg caatgggagg ctgtggagcc ggcgaaataa aatcagagtt 1200gttgcta 12078205PRTHomo sapiens 8Met Ala His Ala Gly Arg Thr Gly Tyr Asp Asn Arg Glu Ile Val Met 1 5 10 15 Lys Tyr Ile His Tyr Lys Leu Ser Gln Arg Gly Tyr Glu Trp Asp Ala 20 25 30 Gly Asp Val Gly Ala Ala Pro Pro Gly Ala Ala Pro Ala Pro Gly Ile 35 40 45 Phe Ser Ser Gln Pro Gly His Thr Pro His Pro Ala Ala Ser Arg Asp 50 55 60 Pro Val Ala Arg Thr Ser Pro Leu Gln Thr Pro Ala Ala Pro Gly Ala 65 70 75 80 Ala Ala Gly Pro Ala Leu Ser Pro Val Pro Pro Val Val His Leu Thr 85 90 95 Leu Arg Gln Ala Gly Asp Asp Phe Ser Arg Arg Tyr Arg Arg Asp Phe 100 105 110 Ala Glu Met Ser Ser Gln Leu His Leu Thr Pro Phe Thr Ala Arg Gly 115 120 125 Arg Phe Ala Thr Val Val Glu Glu Leu Phe Arg Asp Gly Val Asn Trp 130 135 140 Gly Arg Ile Val Ala Phe Phe Glu Phe Gly Gly Val Met Cys Val Glu 145 150 155 160 Ser Val Asn Arg Glu Met Ser Pro Leu Val Asp Asn Ile Ala Leu Trp 165 170 175 Met Thr Glu Tyr Leu Asn Arg His Leu His Thr Trp Ile Gln Asp Asn 180 185 190 Gly Gly Trp Val Gly Ala Leu Gly Asp Val Ser Leu Gly 195 200 205 9

2594DNAHuman immunodeficiency virus 9atggagccag tagatcctag actagagccc tggaagcatc caggaagtca gcctaaaact 60gcttgtacca attgctattg taaaaagtgt tgctttcatt gccaagtttg tttcataaca 120aaagccttag gcatctccta tggcaggaag aagcggagac agcgacgaag agctcatcag 180aacagtcaga ctcatcaagc ttctctatca aagcagtaag tagtacatgt aatgcaacct 240ataccaatag tagcaatagt agcattagta gtagcaataa taatagcaat agttgtgtgg 300tccatagtaa tcatagaata taggaaaata ttaagacaaa gaaaaataga caggttaatt 360gatagactaa tagaaagagc agaagacagt ggcaatgaga gtgaaggaga aatatcagca 420cttgtggaga tgggggtgga gatggggcac catgctcctt gggatgttga tgatctgtag 480tgctacagaa aaattgtggg tcacagtcta ttatggggta cctgtgtgga aggaagcaac 540caccactcta ttttgtgcat cagatgctaa agcatatgat acagaggtac ataatgtttg 600ggccacacat gcctgtgtac ccacagaccc caacccacaa gaagtagtat tggtaaatgt 660gacagaaaat tttaacatgt ggaaaaatga catggtagaa cagatgcatg aggatataat 720cagtttatgg gatcaaagcc taaagccatg tgtaaaatta accccactct gtgttagttt 780aaagtgcact gatttgaaga atgatactaa taccaatagt agtagcggga gaatgataat 840ggagaaagga gagataaaaa actgctcttt caatatcagc acaagcataa gaggtaaggt 900gcagaaagaa tatgcatttt tttataaact tgatataata ccaatagata atgatactac 960cagctataag ttgacaagtt gtaacacctc agtcattaca caggcctgtc caaaggtatc 1020ctttgagcca attcccatac attattgtgc cccggctggt tttgcgattc taaaatgtaa 1080taataagacg ttcaatggaa caggaccatg tacaaatgtc agcacagtac aatgtacaca 1140tggaattagg ccagtagtat caactcaact gctgttaaat ggcagtctag cagaagaaga 1200ggtagtaatt agatctgtca atttcacgga caatgctaaa accataatag tacagctgaa 1260cacatctgta gaaattaatt gtacaagacc caacaacaat acaagaaaaa gaatccgtat 1320ccagagagga ccagggagag catttgttac aataggaaaa ataggaaata tgagacaagc 1380acattgtaac attagtagag caaaatggaa taacacttta aaacagatag ctagcaaatt 1440aagagaacaa tttggaaata ataaaacaat aatctttaag caatcctcag gaggggaccc 1500agaaattgta acgcacagtt ttaattgtgg aggggaattt ttctactgta attcaacaca 1560actgtttaat agtacttggt ttaatagtac ttggagtact gaagggtcaa ataacactga 1620aggaagtgac acaatcaccc tcccatgcag aataaaacaa attataaaca tgtggcagaa 1680agtaggaaaa gcaatgtatg cccctcccat cagtggacaa attagatgtt catcaaatat 1740tacagggctg ctattaacaa gagatggtgg taatagcaac aatgagtccg agatcttcag 1800acctggagga ggagatatga gggacaattg gagaagtgaa ttatataaat ataaagtagt 1860aaaaattgaa ccattaggag tagcacccac caaggcaaag agaagagtgg tgcagagaga 1920aaaaagagca gtgggaatag gagctttgtt ccttgggttc ttgggagcag caggaagcac 1980tatgggcgca gcctcaatga cgctgacggt acaggccaga caattattgt ctggtatagt 2040gcagcagcag aacaatttgc tgagggctat tgaggcgcaa cagcatctgt tgcaactcac 2100agtctggggc atcaagcagc tccaggcaag aatcctggct gtggaaagat acctaaagga 2160tcaacagctc ctggggattt ggggttgctc tggaaaactc atttgcacca ctgctgtgcc 2220ttggaatgct agttggagta ataaatctct ggaacagatt tggaatcaca cgacctggat 2280ggagtgggac agagaaatta acaattacac aagcttaata cactccttaa ttgaagaatc 2340gcaaaaccag caagaaaaga atgaacaaga attattggaa ttagataaat gggcaagttt 2400gtggaattgg tttaacataa caaattggct gtggtatata aaattattca taatgatagt 2460aggaggcttg gtaggtttaa gaatagtttt tgctgtactt tctatagtga atagagttag 2520gcagggatat tcaccattat cgtttcagac ccacctccca accccgaggg gacccgacag 2580gcccgaagga atag 25941086PRTHuman immunodeficiency virus 10Met Glu Pro Val Asp Pro Arg Leu Glu Pro Trp Lys His Pro Gly Ser 1 5 10 15 Gln Pro Lys Thr Ala Cys Thr Asn Cys Tyr Cys Lys Lys Cys Cys Phe 20 25 30 His Cys Gln Val Cys Phe Ile Thr Lys Ala Leu Gly Ile Ser Tyr Gly 35 40 45 Arg Lys Lys Arg Arg Gln Arg Arg Arg Ala His Gln Asn Ser Gln Thr 50 55 60 His Gln Ala Ser Leu Ser Lys Gln Pro Thr Ser Gln Pro Arg Gly Asp 65 70 75 80 Pro Thr Gly Pro Lys Glu 85 112397DNAHomo sapiens 11ctgtcccgca agcgccggcg gcagcatggc cagctctggt tccctgaggg cttcaaagtg 60tctgaggcca gcaagaagaa gcggcgggag cccctcggcg aggactccgt gggcctcaag 120cccctgaaga acgcttcaga cggtgccctc atggacgaca accagaatga gtggggggac 180gaggacctgg agaccaagaa gttccggttc gaggagcccg tggttctgcc tgacctggac 240gaccagacag accaccggca gtggactcag cagcacctgg atgccgctga cctgcgcatg 300tctgccatgg cccccacacc gccccagggt gaggttgacg ccgactgcat ggacgtcaat 360gtccgcgggc ctgatggctt caccccgctc atgatcgcct cctgcagcgg gggcggcctg 420gagacgggca acagcgagga agaggaggac gcgccggccg tcatctccga cttcatctac 480cagggcgcca gcctgcacaa ccagacagac cgcacgggcg agaccgcctt gcacctggcc 540gcccgctact cacgctctga tgccgccaag cgcctgctgg aggccagcgc agatgccaac 600atccaggaca acatgggccg caccccgctg catgcggctg tgtctgccga cgcacaaggt 660gtcttccaga tcctgatccg gaaccgagcc acagacctgg atgcccgcat gcatgatggc 720acgacgccac tgatcctggc tgcccgcctg gccgtggagg gcatgctgga ggacctcatc 780aactcacacg ccgacgtcaa cgccgtagat gacctgggca agtccgccct gcactgggcc 840gccgccgtga acaatgtgga tgccgcagtt gtgctcctga agaacggggc taacaaagat 900atgcagaaca acagggagga gacacccctg tttctggccg cccgggaggg cagctacgag 960accgccaagg tgctgctgga ccactttgcc aaccgggaca tcacggatca tatggaccgc 1020ctgccgcgcg acatcgcaca ggagcgcatg catcacgaca tcgtgaggct gctggacgag 1080tacaacctgg tgcgcagccc gcagctgcac ggagccccgc tggggggcac gcccaccctg 1140tcgcccccgc tctgctcgcc caacggctac ctgggcagcc tcaagcccgg cgtgcagggc 1200aagaaggtcc gcaagcccag cagcaaaggc ctggcctgtg gaagcaagga ggccaaggac 1260ctcaaggcac ggaggaagaa gtcccaggac ggcaagggct gcctgctgga cagctccggc 1320atgctctcgc ccgtggactc cctggagtca ccccatggct acctgtcaga cgtggcctcg 1380ccgccactgc tgccctcccc gttccagcag tctccgtccg tgcccctcaa ccacctgcct 1440gggatgcccg acacccacct gggcatcggg cacctgaacg tggcggccaa gcccgagatg 1500gcggcgctgg gtgggggcgg ccggctggcc tttgagactg gcccacctcg tctctcccac 1560ctgcctgtgg cctctggcac cagcaccgtc ctgggctcca gcagcggagg ggccctgaat 1620ttcactgtgg gcgggtccac cagtttgaat ggtcaatgcg agtggctgtc ccggctgcag 1680agcggcatgg tgccgaacca atacaaccct ctgcggggga gtgtggcacc aggccccctg 1740agcacacagg ccccctccct gcagcatggc atggtaggcc cgctgcacag tagccttgct 1800gccagcgccc tgtcccagat gatgagctac cagggcctgc ccagcacccg gctggccacc 1860cagcctcacc tggtgcagac ccagcaggtg cagccacaaa acttacagat gcagcagcag 1920aacctgcagc cagcaaacat ccagcagcag caaagcctgc agccgccacc accaccacca 1980cagccgcacc ttggcgtgag ctcagcagcc agcggccacc tgggccggag cttcctgagt 2040ggagagccga gccaggcaga cgtgcagcca ctgggcccca gcagcctggc ggtgcacact 2100attctgcccc aggagagccc cgccctgccc acgtcgctgc catcctcgct ggtcccaccc 2160gtgaccgcag cccagttcct gacgcccccc tcgcagcaca gctactcctc gcctgtggac 2220aacaccccca gccaccagct acaggtgcct gagcacccct tcctcacccc gtcccctgag 2280tcccctgacc agtggtccag ctcgtccccg cattccaacg tctccgactg gtccgagggc 2340gtctccagcc ctcccaccag catgcagtcc cagatcgccc gcattccgga ggccttc 239712799PRTHomo sapiens 12Leu Ser Arg Lys Arg Arg Arg Gln His Gly Gln Leu Trp Phe Pro Glu 1 5 10 15 Gly Phe Lys Val Ser Glu Ala Ser Lys Lys Lys Arg Arg Glu Pro Leu 20 25 30 Gly Glu Asp Ser Val Gly Leu Lys Pro Leu Lys Asn Ala Ser Asp Gly 35 40 45 Ala Leu Met Asp Asp Asn Gln Asn Glu Trp Gly Asp Glu Asp Leu Glu 50 55 60 Thr Lys Lys Phe Arg Phe Glu Glu Pro Val Val Leu Pro Asp Leu Asp 65 70 75 80 Asp Gln Thr Asp His Arg Gln Trp Thr Gln Gln His Leu Asp Ala Ala 85 90 95 Asp Leu Arg Met Ser Ala Met Ala Pro Thr Pro Pro Gln Gly Glu Val 100 105 110 Asp Ala Asp Cys Met Asp Val Asn Val Arg Gly Pro Asp Gly Phe Thr 115 120 125 Pro Leu Met Ile Ala Ser Cys Ser Gly Gly Gly Leu Glu Thr Gly Asn 130 135 140 Ser Glu Glu Glu Glu Asp Ala Pro Ala Val Ile Ser Asp Phe Ile Tyr 145 150 155 160 Gln Gly Ala Ser Leu His Asn Gln Thr Asp Arg Thr Gly Glu Thr Ala 165 170 175 Leu His Leu Ala Ala Arg Tyr Ser Arg Ser Asp Ala Ala Lys Arg Leu 180 185 190 Leu Glu Ala Ser Ala Asp Ala Asn Ile Gln Asp Asn Met Gly Arg Thr 195 200 205 Pro Leu His Ala Ala Val Ser Ala Asp Ala Gln Gly Val Phe Gln Ile 210 215 220 Leu Ile Arg Asn Arg Ala Thr Asp Leu Asp Ala Arg Met His Asp Gly 225 230 235 240 Thr Thr Pro Leu Ile Leu Ala Ala Arg Leu Ala Val Glu Gly Met Leu 245 250 255 Glu Asp Leu Ile Asn Ser His Ala Asp Val Asn Ala Val Asp Asp Leu 260 265 270 Gly Lys Ser Ala Leu His Trp Ala Ala Ala Val Asn Asn Val Asp Ala 275 280 285 Ala Val Val Leu Leu Lys Asn Gly Ala Asn Lys Asp Met Gln Asn Asn 290 295 300 Arg Glu Glu Thr Pro Leu Phe Leu Ala Ala Arg Glu Gly Ser Tyr Glu 305 310 315 320 Thr Ala Lys Val Leu Leu Asp His Phe Ala Asn Arg Asp Ile Thr Asp 325 330 335 His Met Asp Arg Leu Pro Arg Asp Ile Ala Gln Glu Arg Met His His 340 345 350 Asp Ile Val Arg Leu Leu Asp Glu Tyr Asn Leu Val Arg Ser Pro Gln 355 360 365 Leu His Gly Ala Pro Leu Gly Gly Thr Pro Thr Leu Ser Pro Pro Leu 370 375 380 Cys Ser Pro Asn Gly Tyr Leu Gly Ser Leu Lys Pro Gly Val Gln Gly 385 390 395 400 Lys Lys Val Arg Lys Pro Ser Ser Lys Gly Leu Ala Cys Gly Ser Lys 405 410 415 Glu Ala Lys Asp Leu Lys Ala Arg Arg Lys Lys Ser Gln Asp Gly Lys 420 425 430 Gly Cys Leu Leu Asp Ser Ser Gly Met Leu Ser Pro Val Asp Ser Leu 435 440 445 Glu Ser Pro His Gly Tyr Leu Ser Asp Val Ala Ser Pro Pro Leu Leu 450 455 460 Pro Ser Pro Phe Gln Gln Ser Pro Ser Val Pro Leu Asn His Leu Pro 465 470 475 480 Gly Met Pro Asp Thr His Leu Gly Ile Gly His Leu Asn Val Ala Ala 485 490 495 Lys Pro Glu Met Ala Ala Leu Gly Gly Gly Gly Arg Leu Ala Phe Glu 500 505 510 Thr Gly Pro Pro Arg Leu Ser His Leu Pro Val Ala Ser Gly Thr Ser 515 520 525 Thr Val Leu Gly Ser Ser Ser Gly Gly Ala Leu Asn Phe Thr Val Gly 530 535 540 Gly Ser Thr Ser Leu Asn Gly Gln Cys Glu Trp Leu Ser Arg Leu Gln 545 550 555 560 Ser Gly Met Val Pro Asn Gln Tyr Asn Pro Leu Arg Gly Ser Val Ala 565 570 575 Pro Gly Pro Leu Ser Thr Gln Ala Pro Ser Leu Gln His Gly Met Val 580 585 590 Gly Pro Leu His Ser Ser Leu Ala Ala Ser Ala Leu Ser Gln Met Met 595 600 605 Ser Tyr Gln Gly Leu Pro Ser Thr Arg Leu Ala Thr Gln Pro His Leu 610 615 620 Val Gln Thr Gln Gln Val Gln Pro Gln Asn Leu Gln Met Gln Gln Gln 625 630 635 640 Asn Leu Gln Pro Ala Asn Ile Gln Gln Gln Gln Ser Leu Gln Pro Pro 645 650 655 Pro Pro Pro Pro Gln Pro His Leu Gly Val Ser Ser Ala Ala Ser Gly 660 665 670 His Leu Gly Arg Ser Phe Leu Ser Gly Glu Pro Ser Gln Ala Asp Val 675 680 685 Gln Pro Leu Gly Pro Ser Ser Leu Ala Val His Thr Ile Leu Pro Gln 690 695 700 Glu Ser Pro Ala Leu Pro Thr Ser Leu Pro Ser Ser Leu Val Pro Pro 705 710 715 720 Val Thr Ala Ala Gln Phe Leu Thr Pro Pro Ser Gln His Ser Tyr Ser 725 730 735 Ser Pro Val Asp Asn Thr Pro Ser His Gln Leu Gln Val Pro Glu His 740 745 750 Pro Phe Leu Thr Pro Ser Pro Glu Ser Pro Asp Gln Trp Ser Ser Ser 755 760 765 Ser Pro His Ser Asn Val Ser Asp Trp Ser Glu Gly Val Ser Ser Pro 770 775 780 Pro Thr Ser Met Gln Ser Gln Ile Ala Arg Ile Pro Glu Ala Phe 785 790 795 13814DNAHomo sapiens 13ccttcacctg cctctgccgc ccaggctaca cgggccacca ctgcgagacc aacatcaacg 60agtgctccag ccagccctgc cgcctacggg gcacctgcca ggacccggac aacgcctacc 120tctgcttctg cctgaagggg accacaggac ccaactgcga gatcaacctg gatgactgtg 180ccagcagccc ctgcgactcg ggcacctgtc tggacaagat cgatggctac gagtgtgcct 240gtgagccggg ctacacaggg agcatgtgta acagcaacat cgatgagtgt gcgggcaacc 300cctgccacaa cgggggcacc tgcgaggacg gcatcaatgg cttcacctgc cgctgccccg 360agggctacca cgaccccacc tgcctgtctg aggtcaatga gtgcaacagc aacccctgcg 420tccacggggc ctgccgggac agcctcaacg ggtacaagtg cgactgtgac cctgggtgga 480gtgggaccaa ctgtgacatc aacaacaacg agtgtgaatc caacccttgt gtcaacggcg 540gcacctgcaa agacatgacc agtggcatcg tgtgcacctg ccgggagggc ttcagcggtc 600ccaactgcca gaccaacatc aacgagtgtg cgtccaaccc atgtctgaac aagggcacgt 660gtattgacga cgttgccggg tacaagtgca actgcctgct gccctacaca ggtgccacgt 720gtgaggtggt gctggccccg tgtgccccca gcccctgcag aaacggcggg gagtgcaggc 780aatccgagga ctatgagagt tgtcactatg tcct 81414799PRTHomo sapiens 14Leu Ser Arg Lys Arg Arg Arg Gln His Gly Gln Leu Trp Phe Pro Glu 1 5 10 15 Gly Phe Lys Val Ser Glu Ala Ser Lys Lys Lys Arg Arg Glu Pro Leu 20 25 30 Gly Glu Asp Ser Val Gly Leu Lys Pro Leu Lys Asn Ala Ser Asp Gly 35 40 45 Ala Leu Met Asp Asp Asn Gln Asn Glu Trp Gly Asp Glu Asp Leu Glu 50 55 60 Thr Lys Lys Phe Arg Phe Glu Glu Pro Val Val Leu Pro Asp Leu Asp 65 70 75 80 Asp Gln Thr Asp His Arg Gln Trp Thr Gln Gln His Leu Asp Ala Ala 85 90 95 Asp Leu Arg Met Ser Ala Met Ala Pro Thr Pro Pro Gln Gly Glu Val 100 105 110 Asp Ala Asp Cys Met Asp Val Asn Val Arg Gly Pro Asp Gly Phe Thr 115 120 125 Pro Leu Met Ile Ala Ser Cys Ser Gly Gly Gly Leu Glu Thr Gly Asn 130 135 140 Ser Glu Glu Glu Glu Asp Ala Pro Ala Val Ile Ser Asp Phe Ile Tyr 145 150 155 160 Gln Gly Ala Ser Leu His Asn Gln Thr Asp Arg Thr Gly Glu Thr Ala 165 170 175 Leu His Leu Ala Ala Arg Tyr Ser Arg Ser Asp Ala Ala Lys Arg Leu 180 185 190 Leu Glu Ala Ser Ala Asp Ala Asn Ile Gln Asp Asn Met Gly Arg Thr 195 200 205 Pro Leu His Ala Ala Val Ser Ala Asp Ala Gln Gly Val Phe Gln Ile 210 215 220 Leu Ile Arg Asn Arg Ala Thr Asp Leu Asp Ala Arg Met His Asp Gly 225 230 235 240 Thr Thr Pro Leu Ile Leu Ala Ala Arg Leu Ala Val Glu Gly Met Leu 245 250 255 Glu Asp Leu Ile Asn Ser His Ala Asp Val Asn Ala Val Asp Asp Leu 260 265 270 Gly Lys Ser Ala Leu His Trp Ala Ala Ala Val Asn Asn Val Asp Ala 275 280 285 Ala Val Val Leu Leu Lys Asn Gly Ala Asn Lys Asp Met Gln Asn Asn 290 295 300 Arg Glu Glu Thr Pro Leu Phe Leu Ala Ala Arg Glu Gly Ser Tyr Glu 305 310 315 320 Thr Ala Lys Val Leu Leu Asp His Phe Ala Asn Arg Asp Ile Thr Asp 325 330 335 His Met Asp Arg Leu Pro Arg Asp Ile Ala Gln Glu Arg Met His His 340 345 350 Asp Ile Val Arg Leu Leu Asp Glu Tyr Asn Leu Val Arg Ser Pro Gln 355 360 365 Leu His Gly Ala Pro Leu Gly Gly Thr Pro Thr Leu Ser Pro Pro Leu 370 375 380 Cys Ser Pro Asn Gly Tyr Leu Gly Ser Leu Lys Pro Gly Val Gln Gly 385 390 395 400 Lys Lys Val Arg Lys Pro Ser Ser Lys Gly Leu Ala Cys Gly Ser Lys 405 410 415 Glu Ala Lys Asp Leu Lys Ala Arg Arg Lys Lys Ser Gln Asp Gly Lys 420 425 430 Gly Cys Leu Leu Asp Ser Ser Gly Met Leu Ser Pro Val Asp Ser Leu 435 440 445 Glu Ser Pro His Gly Tyr Leu Ser Asp Val Ala Ser Pro Pro Leu Leu 450 455 460 Pro Ser Pro Phe Gln Gln Ser Pro Ser Val Pro Leu Asn His Leu Pro 465 470 475 480 Gly Met Pro Asp Thr His Leu Gly Ile Gly His Leu Asn Val Ala Ala 485 490 495 Lys Pro Glu Met Ala Ala Leu Gly

Gly Gly Gly Arg Leu Ala Phe Glu 500 505 510 Thr Gly Pro Pro Arg Leu Ser His Leu Pro Val Ala Ser Gly Thr Ser 515 520 525 Thr Val Leu Gly Ser Ser Ser Gly Gly Ala Leu Asn Phe Thr Val Gly 530 535 540 Gly Ser Thr Ser Leu Asn Gly Gln Cys Glu Trp Leu Ser Arg Leu Gln 545 550 555 560 Ser Gly Met Val Pro Asn Gln Tyr Asn Pro Leu Arg Gly Ser Val Ala 565 570 575 Pro Gly Pro Leu Ser Thr Gln Ala Pro Ser Leu Gln His Gly Met Val 580 585 590 Gly Pro Leu His Ser Ser Leu Ala Ala Ser Ala Leu Ser Gln Met Met 595 600 605 Ser Tyr Gln Gly Leu Pro Ser Thr Arg Leu Ala Thr Gln Pro His Leu 610 615 620 Val Gln Thr Gln Gln Val Gln Pro Gln Asn Leu Gln Met Gln Gln Gln 625 630 635 640 Asn Leu Gln Pro Ala Asn Ile Gln Gln Gln Gln Ser Leu Gln Pro Pro 645 650 655 Pro Pro Pro Pro Gln Pro His Leu Gly Val Ser Ser Ala Ala Ser Gly 660 665 670 His Leu Gly Arg Ser Phe Leu Ser Gly Glu Pro Ser Gln Ala Asp Val 675 680 685 Gln Pro Leu Gly Pro Ser Ser Leu Ala Val His Thr Ile Leu Pro Gln 690 695 700 Glu Ser Pro Ala Leu Pro Thr Ser Leu Pro Ser Ser Leu Val Pro Pro 705 710 715 720 Val Thr Ala Ala Gln Phe Leu Thr Pro Pro Ser Gln His Ser Tyr Ser 725 730 735 Ser Pro Val Asp Asn Thr Pro Ser His Gln Leu Gln Val Pro Glu His 740 745 750 Pro Phe Leu Thr Pro Ser Pro Glu Ser Pro Asp Gln Trp Ser Ser Ser 755 760 765 Ser Pro His Ser Asn Val Ser Asp Trp Ser Glu Gly Val Ser Ser Pro 770 775 780 Pro Thr Ser Met Gln Ser Gln Ile Ala Arg Ile Pro Glu Ala Phe 785 790 795

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed